1
|
Becker IC, Wilkie AR, Nikols E, Carminita E, Roweth HG, Tilburg J, Sciaudone AR, Noetzli LJ, Fatima F, Couldwell G, Ray A, Mogilner A, Machlus KR, Italiano JE. Cell cycle-dependent centrosome clustering precedes proplatelet formation. SCIENCE ADVANCES 2024; 10:eadl6153. [PMID: 38896608 PMCID: PMC11186502 DOI: 10.1126/sciadv.adl6153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Platelet-producing megakaryocytes (MKs) primarily reside in the bone marrow, where they duplicate their DNA content with each cell cycle resulting in polyploid cells with an intricate demarcation membrane system. While key elements of the cytoskeletal reorganizations during proplatelet formation have been identified, what initiates the release of platelets into vessel sinusoids remains largely elusive. Using a cell cycle indicator, we observed a unique phenomenon, during which amplified centrosomes in MKs underwent clustering following mitosis, closely followed by proplatelet formation, which exclusively occurred in G1 of interphase. Forced cell cycle arrest in G1 increased proplatelet formation not only in vitro but also in vivo following short-term starvation of mice. We identified that inhibition of the centrosomal protein kinesin family member C1 (KIFC1) impaired clustering and subsequent proplatelet formation, while KIFC1-deficient mice exhibited reduced platelet counts. In summary, we identified KIFC1- and cell cycle-mediated centrosome clustering as an important initiator of proplatelet formation from MKs.
Collapse
Affiliation(s)
- Isabelle C. Becker
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Adrian R. Wilkie
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Emma Nikols
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
| | - Estelle Carminita
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Harvey G. Roweth
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Brigham and Women’s Hospital, 4 Blackfan Circle, Boston, MA 02115, USA
| | - Julia Tilburg
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | | | - Leila J. Noetzli
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Brigham and Women’s Hospital, 4 Blackfan Circle, Boston, MA 02115, USA
| | - Farheen Fatima
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
| | | | - Anjana Ray
- Brigham and Women’s Hospital, 4 Blackfan Circle, Boston, MA 02115, USA
| | - Alex Mogilner
- Courant Institute of Mathematical Sciences, New York University, 251 Mercer Street, New York, NY 10012, USA
| | - Kellie R. Machlus
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Joseph E. Italiano
- Vascular Biology Program, Boston Children’s Hospital, 1 Blackfan Circle, Boston, MA 02115, USA
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| |
Collapse
|
2
|
Si LH, Sun GC, Liu ZW, Gu SY, Yan CH, Xu JY, Jia Y. High expression levels of centromere protein O participates in cell proliferation of human ovarian cancer. J Ovarian Res 2024; 17:126. [PMID: 38890751 PMCID: PMC11184697 DOI: 10.1186/s13048-024-01452-x] [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: 03/13/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
Ovarian cancer is a common malignant tumor in women, with a high mortality rate ranking first among gynecological tumors. Currently, there is insufficient understanding of the causes, pathogenesis, recurrence and metastasis of ovarian cancer, and early diagnosis and treatment still face great challenges. The sensitivity and specificity of existing ovarian cancer screening methods are still unsatisfactory. Centromere protein O (CENP-O) is a recently discovered structural centromere protein that is involved in cell death and is essential for spindle assembly, chromosome separation, and checkpoint signaling during mitosis. The abnormal high expression of CENP-O was detected in various tumors such as bladder cancer and gastric cancer, and it participates in the regulation of tumor cell proliferation. In this study, we detect the expression abundance of CENP-O mRNA in different ovarian cancer cells ( ES-2, A2780, Caov-3, OVCAR-3 and SK-OV-3). The biological function changes of cell proliferation and apoptosis were detected and the role of CENP-O in ovarian cancer cell proliferation and apoptosis was explored by knocking down the expression of CENP-O gene. The results showed that CENP-O gene was significantly expressed in 5 types of ovarian cancer cell lines. After knocking down the CENP-O gene, the proliferation and cloning ability of ovarian cancer cells decreased, and the apoptosis increased. This study indicates that CENP-O has the potential to be a molecular therapeutic target, and downregulating the expression of CENP-O gene can break the unlimited proliferation ability of cancer cells and promote their apoptosis, providing a foundation and new ideas for subsequent molecular mechanism research and targeted therapy.
Collapse
Affiliation(s)
- Li-Hui Si
- Department of Obstetrics and Gynecology, The second Hospital of Jilin University, Changchun, Jilin, China
| | - Guang-Chao Sun
- Department of Neurosurgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Zi-Wei Liu
- Department of Obstetrics and Gynecology, The second Hospital of Jilin University, Changchun, Jilin, China
| | - Shi-Yu Gu
- Department of Obstetrics and Gynecology, The second Hospital of Jilin University, Changchun, Jilin, China
| | - Chu-Han Yan
- Department of Obstetrics and Gynecology, The second Hospital of Jilin University, Changchun, Jilin, China
| | - Jin-Yuan Xu
- Department of Obstetrics and Gynecology, The second Hospital of Jilin University, Changchun, Jilin, China
| | - Yan Jia
- Department of Obstetrics and Gynecology, The second Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
3
|
Banerjee DS, Banerjee S. Catalytic growth in a shared enzyme pool ensures robust control of centrosome size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543875. [PMID: 37333186 PMCID: PMC10274694 DOI: 10.1101/2023.06.06.543875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Accurate regulation of centrosome size is essential for ensuring error-free cell division, and dysregulation of centrosome size has been linked to various pathologies, including developmental defects and cancer. While a universally accepted model for centrosome size regulation is lacking, prior theoretical and experimental work suggest a centrosome growth model involving autocatalytic assembly of the pericentriolic material. Here we show that the autocatalytic assembly model fails to explain the attainment of equal centrosome sizes, which is crucial for error-free cell division. Incorporating latest experimental findings into the molecular mechanisms governing centrosome assembly, we introduce a new quantitative theory for centrosome growth involving catalytic assembly within a shared pool of enzymes. Our model successfully achieves robust size equality between maturing centrosome pairs, mirroring cooperative growth dynamics observed in experiments. To validate our theoretical predictions, we compare them with available experimental data and demonstrate the broad applicability of the catalytic growth model across different organisms, which exhibit distinct growth dynamics and size scaling characteristics.
Collapse
Affiliation(s)
- Deb Sankar Banerjee
- Department of Physics, Carnegie Mellon University, Pittsburgh, PA 15213, USA
- James Franck Institute, University of Chicago, Chicago, IL 60637, USA
| | - Shiladitya Banerjee
- Department of Physics, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
4
|
Tingler M, Philipp M, Burkhalter MD. DNA Replication proteins in primary microcephaly syndromes. Biol Cell 2022; 114:143-159. [PMID: 35182397 DOI: 10.1111/boc.202100061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 11/30/2022]
Abstract
SCOPE Improper expansion of neural stem and progenitor cells during brain development manifests in primary microcephaly. It is characterized by a reduced head circumference, which correlates with a reduction in brain size. This often corresponds to a general underdevelopment of the brain and entails cognitive, behavioral and motoric retardation. In the past decade significant research efforts have been undertaken to identify genes and the molecular mechanisms underlying microcephaly. One such gene set encompasses factors required for DNA replication. Intriguingly, a growing body of evidence indicates that a substantial number of these genes mediate faithful centrosome and cilium function in addition to their canonical function in genome duplication. Here, we summarize, which DNA replication factors are associated with microcephaly syndromes and to which extent they impact on centrosomes and cilia. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Melanie Tingler
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University Tübingen, Tübingen, 72074, Germany
| | - Melanie Philipp
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University Tübingen, Tübingen, 72074, Germany
| | - Martin D Burkhalter
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, Section of Pharmacogenomics, Eberhard-Karls-University Tübingen, Tübingen, 72074, Germany
| |
Collapse
|
5
|
The potential role of COVID-19 in the induction of DNA damage. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2022; 789:108411. [PMID: 35690420 PMCID: PMC8767986 DOI: 10.1016/j.mrrev.2022.108411] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 11/30/2021] [Accepted: 01/17/2022] [Indexed: 01/07/2023]
Abstract
The coronavirus disease-2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is challenging global health and economic systems. In some individuals, COVID-19 can cause a wide array of symptoms, affecting several organs, such as the lungs, heart, bowels, kidneys and brain, causing multiorgan failure, sepsis and death. These effects are related in part to direct viral infection of these organs, immunological deregulation, a hypercoagulatory state and the potential for development of cytokine storm syndrome. Since the appearance of COVID-19 is recent, the long-term effects on the health of recovered patients remain unknown. In this review, we focused on current evidence of the mechanisms of DNA damage mediated by coronaviruses. Data supports that these viruses can induce DNA damage, genomic instability, and cell cycle deregulation during their replication in mammalian cells. Since the induction of DNA damage and aberrant DNA repair mechanisms are related to the development of chronic diseases such as cancer, diabetes, neurodegenerative disorders, and atherosclerosis, it will be important to address similar effects and outcomes in recovered COVID-19 patients.
Collapse
|
6
|
Zhou H, Bian T, Qian L, Zhao C, Zhang W, Zheng M, Zhou H, Liu L, Sun H, Li X, Zhang J, Liu Y. Prognostic model of lung adenocarcinoma constructed by the CENPA complex genes is closely related to immune infiltration. Pathol Res Pract 2021; 228:153680. [PMID: 34798483 DOI: 10.1016/j.prp.2021.153680] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is still one of the primary malignant diseases leading to higher mortality worldwide. It has been previously reported that multiple genes in the CENPA-nucleosome associated complex (NAC) complex in lung cancer can be used as prognostic markers; however, there is lack of comprehensive research on the CENPA-NAC complex. METHODS The hub genes of lung cancer were obtained by analyzing multiple gene expression omnibus (GEO) lung cancer datasets. The key genes of the CENPA-NAC complex in the evolution of LUAD were identified according to lung cancer data obtained from The Cancer Genome Atlas (TCGA) database, and the key genes were constructed as a survival prognostic model. The relationship between the model and immune cell infiltration was studied by the Tumor Immune Estimation Resource (TIMER) and single-sample gene set enrichment analysis (ssGSEA) studies.Droplet Digital polymerase chain reaction (ddPCR) was used to verify the effectiveness of the prognostic model to predict survival using clinical samples. RESULTS A comprehensive study showed that CENPA, CENPH, CENPM, CENPN and CENPU were key genes in the development and evolution of LUAD. The constructed survival prognosis model was an independent risk factor for LUAD and can be used to assess the survival of LUAD patients. The risk score was closely related to the infiltration of multiple immune cells. The independent cohorts GSE31210 and GSE50081 further confirmed the validity of the prognostic model, and finally, the model was validated with clinical samples. CONCLUSIONS In conclusion, the results of the present study showed that CENPA, CENPH, CENPM, CENPN, and CENPU are a group of potential prognostic markers in LUAD. The constructed model has been confirmed to be applicable in the clinical setting in evaluating the survival of patients with LUAD, and providing more evidence on immunotherapy for LUAD.
Collapse
Affiliation(s)
- Haomiao Zhou
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China; Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Tingting Bian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Li Qian
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Cui Zhao
- Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Weiju Zhang
- Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Miaosen Zheng
- Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Hao Zhou
- Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Lei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Hui Sun
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Xiaoli Li
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China
| | - Jianguo Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China; Medical School of Nantong University, Nantong 226001, Jiangsu, China.
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China; Medical School of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
7
|
Beta-Genus Human Papillomavirus 8 E6 Destabilizes the Host Genome by Promoting p300 Degradation. Viruses 2021; 13:v13081662. [PMID: 34452526 PMCID: PMC8402844 DOI: 10.3390/v13081662] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 01/10/2023] Open
Abstract
The beta genus of human papillomaviruses infects cutaneous keratinocytes. Their replication depends on actively proliferating cells and, thus, they conflict with the cellular response to the DNA damage frequently encountered by these cells. This review focus on one of these viruses (HPV8) that counters the cellular response to damaged DNA and mitotic errors by expressing a protein (HPV8 E6) that destabilizes a histone acetyltransferase, p300. The loss of p300 results in broad dysregulation of cell signaling that decreases genome stability. In addition to discussing phenotypes caused by p300 destabilization, the review contains a discussion of the extent to which E6 from other β-HPVs destabilizes p300, and provides a discussion on dissecting HPV8 E6 biology using mutants.
Collapse
|
8
|
Kawakami M, Mustachio LM, Chen Y, Chen Z, Liu X, Wei CH, Roszik J, Kittai AS, Danilov AV, Zhang X, Fang B, Wang J, Heymach JV, Tyutyunyk-Massey L, Freemantle SJ, Kurie JM, Liu X, Dmitrovsky E. A Novel CDK2/9 Inhibitor CYC065 Causes Anaphase Catastrophe and Represses Proliferation, Tumorigenesis, and Metastasis in Aneuploid Cancers. Mol Cancer Ther 2020; 20:477-489. [PMID: 33277443 DOI: 10.1158/1535-7163.mct-19-0987] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 06/18/2020] [Accepted: 11/30/2020] [Indexed: 12/16/2022]
Abstract
Cyclin-dependent kinase 2 (CDK2) antagonism inhibits clustering of excessive centrosomes at mitosis, causing multipolar cell division and apoptotic death. This is called anaphase catastrophe. To establish induced anaphase catastrophe as a clinically tractable antineoplastic mechanism, induced anaphase catastrophe was explored in different aneuploid cancers after treatment with CYC065 (Cyclacel), a CDK2/9 inhibitor. Antineoplastic activity was studied in preclinical models. CYC065 treatment augmented anaphase catastrophe in diverse cancers including lymphoma, lung, colon, and pancreatic cancers, despite KRAS oncoprotein expression. Anaphase catastrophe was a broadly active antineoplastic mechanism. Reverse phase protein arrays (RPPAs) revealed that along with known CDK2/9 targets, focal adhesion kinase and Src phosphorylation that regulate metastasis were each repressed by CYC065 treatment. Intriguingly, CYC065 treatment decreased lung cancer metastases in in vivo murine models. CYC065 treatment also significantly reduced the rate of lung cancer growth in syngeneic murine and patient-derived xenograft (PDX) models independent of KRAS oncoprotein expression. Immunohistochemistry analysis of CYC065-treated lung cancer PDX models confirmed repression of proteins highlighted by RPPAs, implicating them as indicators of CYC065 antitumor response. Phospho-histone H3 staining detected anaphase catastrophe in CYC065-treated PDXs. Thus, induced anaphase catastrophe after CYC065 treatment can combat aneuploid cancers despite KRAS oncoprotein expression. These findings should guide future trials of this novel CDK2/9 inhibitor in the cancer clinic.
Collapse
Affiliation(s)
- Masanori Kawakami
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Lisa Maria Mustachio
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yulong Chen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zibo Chen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Xiuxia Liu
- Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Cheng-Hsin Wei
- Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jason Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adam S Kittai
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Alexey V Danilov
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon
| | - Xiaoshan Zhang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Jonathan M Kurie
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xi Liu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Ethan Dmitrovsky
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Frederick National Laboratory for Cancer Research, Frederick, Maryland.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
9
|
Youn Y, Lee JC, Kim J, Kim JH, Hwang JH. Cdc6 disruption leads to centrosome abnormalities and chromosome instability in pancreatic cancer cells. Sci Rep 2020; 10:16518. [PMID: 33020506 PMCID: PMC7536414 DOI: 10.1038/s41598-020-73474-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Cell division cycle 6 (Cdc6) plays key roles in regulating DNA replication, and activation and maintenance of cell cycle check points. In addition, Cdc6 exerts oncogenic properties via genomic instability associated with incomplete DNA replication. This study aimed to examine the effects of Cdc6 on pancreatic cancer (PC) cells. Our results showed that Cdc6 expression was higher in clinical PC specimens (based on analysis of the GEPIA database) and cell lines, and the high Cdc6 expression was associated with poorer survival in The Cancer Genome Atlas-PC cohort. In addition, Cdc6-depleted PC cells significantly inhibited cell proliferation and colony formation, delayed G2/M cell cycle progression, and increased expression of p-histone H3 and cyclin A2 levels. These observations could be explained by Cdc6 depletion leading to multipolar and split spindles via centrosome amplification and microtubule disorganization which eventually increases chromosome missegregation. Furthermore, Cdc6-depleted PC cells showed significantly increased apoptosis, which was consistent with increased caspase-9 and caspase-3 activation. Collectively, our results demonstrated that Cdc6-depleted PC cells are arrested in mitosis and eventually undergo cell death by induced multipolar spindles, centrosome aberrations, microtubule disorganization, and chromosome instability. In conclusion, Cdc6 may be a potential biomarker and therapeutic target for PC.
Collapse
Affiliation(s)
- Yuna Youn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jae Hyeong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea.
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea. .,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
10
|
CEP131 Abrogates CHK1 Inhibitor-Induced Replication Defects and Is Associated with Unfavorable Outcome in Neuroblastoma. JOURNAL OF ONCOLOGY 2020; 2020:2752417. [PMID: 33014050 PMCID: PMC7512061 DOI: 10.1155/2020/2752417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/11/2020] [Accepted: 08/25/2020] [Indexed: 12/16/2022]
Abstract
Checkpoint kinase 1 (CHK1) plays a key role in genome surveillance and integrity throughout the cell cycle. Selective inhibitors of CHK1 (CHK1i) are undergoing clinical evaluation for various human malignancies, including neuroblastoma. Recently, we reported that CHK1i, PF-477736, induced a p53-mediated DNA damage response. As a result, the cancer cells were able to repair DNA damage and became less sensitive to CHK1i. In this study, we discovered that PF-477736 increased expression of MDM2 oncogene along with CHK1i-induced replication defects in neuroblastoma NB-39-nu cells. A mass spectrometry analysis of protein binding to MDM2 in the presence of CHK1i identified the centrosome-associated family protein 131 (CEP131), which was correlated with unfavorable prognosis of neuroblastoma patients. We revealed that MDM2 was associated with CEP131 protein degradation, whereas overexpression of CEP131 accelerated neuroblastoma cell growth and exhibited resistance to CHK1i-induced replication defects. Thus, these findings may provide a future therapeutic strategy against centrosome-associated oncogenes involving CEP131 as a target in neuroblastoma.
Collapse
|
11
|
Bilinski M, Lanari C, Fabris VT. Centrosome Abnormalities and Polyploidy in Murine Mammary Carcinomas with Different Degrees of Hormone Responsiveness. Cancer Invest 2020; 38:300-309. [PMID: 32378982 DOI: 10.1080/07357907.2020.1766482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Centrosome amplification leads to aberrant mitosis, giving rise to aneuploidy and it has been associated with poor prognosis in human cancers. This study aimed to evaluate the relationship between polyploidy, centrosome abnormalities, and response to endocrine treatment in progestin-induced mouse mammary carcinomas. We found cells with three or more centrosomes in the polyploid tumors. The endocrine unresponsive tumors showed a higher average number of centrosomes per cell than the responsive tumors. The results suggest an association between polyploidy and centrosome amplification with the resistance to endocrine therapy in this luminal breast cancer model.
Collapse
Affiliation(s)
- Melina Bilinski
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Claudia Lanari
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Victoria T Fabris
- Laboratory of Hormonal Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| |
Collapse
|
12
|
Liu C, Zhao J, Lu W, Dai Y, Hockings J, Zhou Y, Nussinov R, Eng C, Cheng F. Individualized genetic network analysis reveals new therapeutic vulnerabilities in 6,700 cancer genomes. PLoS Comput Biol 2020; 16:e1007701. [PMID: 32101536 PMCID: PMC7062285 DOI: 10.1371/journal.pcbi.1007701] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 03/09/2020] [Accepted: 01/30/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor-specific genomic alterations allow systematic identification of genetic interactions that promote tumorigenesis and tumor vulnerabilities, offering novel strategies for development of targeted therapies for individual patients. We develop an Individualized Network-based Co-Mutation (INCM) methodology by inspecting over 2.5 million nonsynonymous somatic mutations derived from 6,789 tumor exomes across 14 cancer types from The Cancer Genome Atlas. Our INCM analysis reveals a higher genetic interaction burden on the significantly mutated genes, experimentally validated cancer genes, chromosome regulatory factors, and DNA damage repair genes, as compared to human pan-cancer essential genes identified by CRISPR-Cas9 screenings on 324 cancer cell lines. We find that genes involved in the cancer type-specific genetic subnetworks identified by INCM are significantly enriched in established cancer pathways, and the INCM-inferred putative genetic interactions are correlated with patient survival. By analyzing drug pharmacogenomics profiles from the Genomics of Drug Sensitivity in Cancer database, we show that the network-predicted putative genetic interactions (e.g., BRCA2-TP53) are significantly correlated with sensitivity/resistance of multiple therapeutic agents. We experimentally validated that afatinib has the strongest cytotoxic activity on BT474 (IC50 = 55.5 nM, BRCA2 and TP53 co-mutant) compared to MCF7 (IC50 = 7.7 μM, both BRCA2 and TP53 wild type) and MDA-MB-231 (IC50 = 7.9 μM, BRCA2 wild type but TP53 mutant). Finally, drug-target network analysis reveals several potential druggable genetic interactions by targeting tumor vulnerabilities. This study offers a powerful network-based methodology for identification of candidate therapeutic pathways that target tumor vulnerabilities and prioritization of potential pharmacogenomics biomarkers for development of personalized cancer medicine. Recent efforts to map genetic interactions in tumor cells have suggested that tumor vulnerabilities can be exploited for development of novel targeted therapies. Tumor-specific genomic alterations derived from multi-center cancer genome projects allow identification of genetic interactions that promote tumor vulnerabilities, offering novel strategies for development of targeted cancer therapies. This study develops a novel Individualized Network-based Co-Mutation (termed INCM) methodology for quantifying the putative genetic interactions in cancer. Trained on over 2.5 million nonsynonymous somatic mutations derived from 6,789 tumor exomes across 14 cancer type, we found that genes identified in the cancer type-specific genetic subnetworks were significantly enriched in established cancer pathways. The network-predicted putative genetic interactions are correlated with patient survival. By analyzing drug pharmacogenomics profiles, we showed that the network-predicted putative genetic interactions (e.g., BRCA2-TP53) were significantly correlated with sensitivity/resistance of anticancer drugs (e.g., afatinib) and we experimentally validated it in breast cancer cell lines. Finally, drug-target network analysis reveals several potential druggable genetic interactions (e.g., PIK3CA-PTEN) by targeting tumor vulnerabilities. This study offers a generalizable network-based approach for comprehensive identification of candidate therapeutic pathways that target tumor vulnerabilities and prioritization of potential prognostic and pharmacogenomics biomarkers for development of personalized cancer medicine.
Collapse
Affiliation(s)
- Chuang Liu
- Alibaba Research Center for Complexity Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Junfei Zhao
- Department of Systems Biology, Columbia University, New York, New York, United States of America
- Department of Biomedical Informatics, Columbia University, New York, New York, United States of America
| | - Weiqiang Lu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yao Dai
- Alibaba Research Center for Complexity Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Jennifer Hockings
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Yadi Zhou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute at Frederick, Frederick, Maryland, United States of America
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
13
|
Kim DH, Kim HM, Huong PTT, Han HJ, Hwang J, Cha-Molstad H, Lee KH, Ryoo IJ, Kim KE, Huh YH, Ahn JS, Kwon YT, Soung NK, Kim BY. Enhanced anticancer effects of a methylation inhibitor by inhibiting a novel DNMT1 target, CEP 131, in cervical cancer. BMB Rep 2019. [PMID: 31068247 PMCID: PMC6549914 DOI: 10.5483/bmbrep.2019.52.5.055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Methylation is a primary epigenetic mechanism regulating gene expression. 5-aza-2′-deoxycytidine is an FDA-approved drug prescribed for treatment of cancer by inhibiting DNA-Methyl-Transferase 1 (DNMT1). Results of this study suggest that prolonged treatment with 5-aza-2′-deoxycytidine could induce centrosome abnormalities in cancer cells and that CEP131, a centrosome protein, is regulated by DNMT1. Interestingly, cancer cell growth was attenuated in vitro and in vivo by inhibiting the expression of Cep131. Finally, Cep131-deficient cells were more sensitive to treatment with DNMT1 inhibitors. These findings suggest that Cep131 is a potential novel anti-cancer target. Agents that can inhibit this protein may be useful alone or in combination with DNMT1 inhibitors to treat cancer.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116; Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Hye-Min Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Pham Thi Thu Huong
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Ho-Jin Han
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116; Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Joonsung Hwang
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Hyunjoo Cha-Molstad
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Kyung Ho Lee
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - In-Ja Ryoo
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Kyoon Eon Kim
- Department of Biochemistry, College of Natural Science, Chungnam National University, Daejeon 34134, Korea
| | - Yang Hoon Huh
- Center for Electron Microscopy Research, Korea Basic Science Institute, Cheongju 28119, Korea
| | - Jong Seog Ahn
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116; Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Yong Tae Kwon
- Protein Metabolism Medical Research Center, Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Korea
| | - Nak-Kyun Soung
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116; Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Bo Yeon Kim
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116; Department of Bio-Molecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| |
Collapse
|
14
|
Cao Y, Xiong J, Li Z, Zhang G, Tu Y, Wang L, Jie Z. CENPO expression regulates gastric cancer cell proliferation and is associated with poor patient prognosis. Mol Med Rep 2019; 20:3661-3670. [PMID: 31485675 PMCID: PMC6755171 DOI: 10.3892/mmr.2019.10624] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 07/17/2019] [Indexed: 01/01/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide; however, understanding of its development and carcinogenesis is currently limited. Centromere protein O (CENPO), is a newly discovered constitutive centromeric protein, associated with cell death. The expression of CENPO in human cancers, including GC, is currently unknown. The aim of the present study was to investigate the clinical association between CENPO and GC, and to elucidate the potential mechanisms of CENPO in the process of GC progression. The results demonstrated that CENPO was expressed at high levels in GC and was correlated with p-TNM stage. In addition, CENPO was observed to be a marker of poor prognosis in patients with GC. Knockdown of CENPO contributed to GC cell growth inhibition and apoptosis induction. In addition, downregulation of CENPO expression suppressed GC cell growth in vivo. Furthermore, CENPO knockdown decreased ataxia telangiectasia mutated (ATM), cyclin D1 and c-Jun expression, indicating that the ATM signaling pathway may be involved in CENPO-mediated regulation of GC cell growth. In conclusion, increased CENPO expression may be associated with the aggressive tumor biology of GC and CENPO may present a novel therapeutic target and prognostic biomarker for patients with GC.
Collapse
Affiliation(s)
- Yi Cao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jianbo Xiong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhengrong Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Guoyang Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yi Tu
- Department of Pathology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lizhen Wang
- Department of Pathology, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhigang Jie
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
15
|
Huang C, Wang L, Song H, Wu C. Interactive effects of AURKA polymorphisms with smoking on the susceptibility of oral cancer. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2333-2337. [PMID: 31174434 DOI: 10.1080/21691401.2019.1601101] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Chao Huang
- Department of Stomatology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Lili Wang
- Department of Prosthodontics, Tianjin Stomatological Hospital, Tianjin, China
| | - Hongguang Song
- Department of Stomatology, Beijing DCN Orthopaedic Hospital, Beijing, China
| | - Cungang Wu
- Department of Ultrasonography, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
16
|
Kawakami M, Liu X, Dmitrovsky E. New Cell Cycle Inhibitors Target Aneuploidy in Cancer Therapy. Annu Rev Pharmacol Toxicol 2018; 59:361-377. [PMID: 30110577 DOI: 10.1146/annurev-pharmtox-010818-021649] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aneuploidy is a hallmark of cancer. Defects in chromosome segregation result in aneuploidy. Multiple pathways are engaged in this process, including errors in kinetochore-microtubule attachments, supernumerary centrosomes, spindle assembly checkpoint (SAC) defects, and chromosome cohesion defects. Although aneuploidy provides an adaptation and proliferative advantage in affected cells, excessive aneuploidy beyond a critical level can be lethal to cancer cells. Given this, enhanced chromosome missegregation is hypothesized to limit survival of aneuploid cancer cells, especially when compared to diploid cells. Based on this concept, proteins and pathways engaged in chromosome segregation are being exploited as candidate therapeutic targets for aneuploid cancers. Agents that induce chromosome missegregation and aneuploidy now exist, including SAC inhibitors, those that alter centrosome fidelity and others that are under active study in preclinical and clinical contexts. This review explores the therapeutic potentials of such new agents, including the benefits of combining them with other antineoplastic agents.
Collapse
Affiliation(s)
- Masanori Kawakami
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas 77030, USA
| | - Xi Liu
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas 77030, USA
| | - Ethan Dmitrovsky
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, The University of Texas, Houston, Texas 77030, USA.,Department of Cancer Biology, MD Anderson Cancer Center, The University of Texas, Houston, Texas 77030, USA.,Current affiliation: Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, USA;
| |
Collapse
|
17
|
Kawakami M, Mustachio LM, Liu X, Dmitrovsky E. Engaging Anaphase Catastrophe Mechanisms to Eradicate Aneuploid Cancers. Mol Cancer Ther 2018; 17:724-731. [PMID: 29559545 DOI: 10.1158/1535-7163.mct-17-1108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/16/2018] [Accepted: 02/16/2018] [Indexed: 12/18/2022]
Abstract
Cancer cells often have supernumerary centrosomes that promote genomic instability, a pathognomonic feature of cancer. During mitosis, cancer cells with supernumerary centrosomes undergo bipolar cell division by clustering centrosomes into two poles. When supernumerary centrosome clustering is antagonized, cancer cells are forced to undergo multipolar division leading to death of daughter cells. This proapoptotic pathway, called anaphase catastrophe, preferentially eliminates aneuploid cancer cells and malignant tumors in engineered mouse models. Anaphase catastrophe occurs through the loss or inhibition of the centrosomal protein CP110, a direct cyclin-dependent kinase 1 (CDK1) and CDK2 target. Intriguingly, CP110 is repressed by the KRAS oncoprotein. This sensitizes KRAS-driven lung cancers (an unmet medical need) to respond to CDK2 inhibitors. Anaphase catastrophe-inducing agents like CDK1 and CDK2 antagonists are lethal to cancer cells with supernumerary centrosomes, but can relatively spare normal cells with two centrosomes. This mechanism is proposed to provide a therapeutic window in the cancer clinic following treatment with a CDK1 or CDK2 inhibitor. Taken together, anaphase catastrophe is a clinically tractable mechanism that promotes death of neoplastic tumors with aneuploidy, a hallmark of cancer. Mol Cancer Ther; 17(4); 724-31. ©2018 AACR.
Collapse
Affiliation(s)
- Masanori Kawakami
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lisa Maria Mustachio
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xi Liu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ethan Dmitrovsky
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| |
Collapse
|
18
|
Rogne M, Svaerd O, Madsen-Østerbye J, Hashim A, Tjønnfjord GE, Staerk J. Cytokinesis arrest and multiple centrosomes in B cell chronic lymphocytic leukaemia. J Cell Mol Med 2018. [PMID: 29516674 PMCID: PMC5908127 DOI: 10.1111/jcmm.13579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cytokinesis failure leads to the emergence of tetraploid cells and multiple centrosomes. Chronic lymphocytic leukaemia (CLL) is the most common haematological malignancy in adults and is characterized by clonal B cell expansion. Here, we show that a significant number of peripheral blood CLL cells are arrested in cytokinesis and that this event occurred after nuclear envelope reformation and before cytoplasmic abscission. mRNA expression data showed that several genes known to be crucial for cell cycle regulation, checkpoint and centromere function, such as ING4, ING5, CDKN1A and CDK4, were significantly dysregulated in CLL samples. Our results demonstrate that CLL cells exhibit difficulties in completing mitosis, which is different from but may, at least in part, explain the previously reported accumulation of CLL cells in G0/1.
Collapse
Affiliation(s)
- Marie Rogne
- Centre for Molecular Medicine Norway, Nordic European Molecular Laboratory Partnership, University of Oslo, Oslo, Norway
| | - Oksana Svaerd
- Centre for Molecular Medicine Norway, Nordic European Molecular Laboratory Partnership, University of Oslo, Oslo, Norway
| | - Julia Madsen-Østerbye
- Centre for Molecular Medicine Norway, Nordic European Molecular Laboratory Partnership, University of Oslo, Oslo, Norway
| | - Adnan Hashim
- Centre for Molecular Medicine Norway, Nordic European Molecular Laboratory Partnership, University of Oslo, Oslo, Norway
| | - Geir E Tjønnfjord
- Department of Haematology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Judith Staerk
- Centre for Molecular Medicine Norway, Nordic European Molecular Laboratory Partnership, University of Oslo, Oslo, Norway.,Department of Haematology, Oslo University Hospital, Oslo, Norway.,Norwegian Center for Stem Cell Research, Department of Immunology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
19
|
Nguyen N, Kumar A, Chacko S, Ouellette RJ, Ghosh A. Human hyaluronic acid synthase-1 promotes malignant transformation via epithelial-to-mesenchymal transition, micronucleation and centrosome abnormalities. Cell Commun Signal 2017; 15:48. [PMID: 29137675 PMCID: PMC5686803 DOI: 10.1186/s12964-017-0204-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/06/2017] [Indexed: 01/25/2023] Open
Abstract
Background Human hyaluronic acid (HA) molecules are synthesized by three membrane spanning Hyaluronic Acid Synthases (HAS1, HAS2 and HAS3). Of the three, HAS1 is found to be localized more into the cytoplasmic space where it synthesizes intracellular HA. HA is a ubiquitous glycosaminoglycan, mainly present in the extracellular matrix (ECM) and on the cell surface, but are also detected intracellularly. Accumulation of HA in cancer cells, the cancer-surrounding stroma, and ECM is generally considered an independent prognostic factors for patients. Higher HA production also correlates with higher tumor grade and more genetic heterogeneity in multiple cancer types which is known to contribute to drug resistance and results in treatment failure. Tumor heterogeneity and intra-tumor clonal diversity are major challenges for diagnosis and treatment. Identification of the driver pathway(s) that initiate genomic instability, tumor heterogeneity and subsequent phenotypic/clinical manifestations, are fundamental for the diagnosis and treatment of cancer. Thus far, no evidence was shown to correlate intracellular HA status (produced by HAS1) and the generation of genetic diversity in tumors. Methods We tested different cell lines engineered to induce HAS1 expression. We measured the epithelial traits, centrosomal abnormalities, micronucleation and polynucleation of those HAS1-expressing cells. We performed real-time PCR, 3D cell culture assay, confocal microscopy, immunoblots and HA-capture methods. Results Our results demonstrate that overexpression of HAS1 induces loss of epithelial traits, increases centrosomal abnormalities, micronucleation and polynucleation, which together indicate manifestation of malignant transformation, intratumoral genetic heterogeneity, and possibly create suitable niche for cancer stem cells generation. Conclusions The intracellular HA produced by HAS1 can aggravate genomic instability and intratumor heterogeneity, pointing to a fundamental role of intracellular HA in cancer initiation and progression. Electronic supplementary material The online version of this article (10.1186/s12964-017-0204-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nguyet Nguyen
- Atlantic Cancer Research Institute, 35 Providence Street, Moncton, NB, E1C 8X3, Canada
| | - Awanit Kumar
- Atlantic Cancer Research Institute, 35 Providence Street, Moncton, NB, E1C 8X3, Canada
| | - Simi Chacko
- Atlantic Cancer Research Institute, 35 Providence Street, Moncton, NB, E1C 8X3, Canada
| | - Rodney J Ouellette
- Atlantic Cancer Research Institute, 35 Providence Street, Moncton, NB, E1C 8X3, Canada.,Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada
| | - Anirban Ghosh
- Atlantic Cancer Research Institute, 35 Providence Street, Moncton, NB, E1C 8X3, Canada. .,Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, Canada.
| |
Collapse
|
20
|
Lee JC, Lu TP, Changou CA, Liang CW, Huang HN, Lauria A, Huang HY, Lin CY, Chiang YC, Davidson B, Lin MC, Kuo KT. Genomewide copy number analysis of Müllerian adenosarcoma identified chromosomal instability in the aggressive subgroup. Mod Pathol 2016; 29:1070-82. [PMID: 27255164 DOI: 10.1038/modpathol.2016.99] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 04/23/2016] [Accepted: 04/24/2016] [Indexed: 12/19/2022]
Abstract
Müllerian adenosarcomas are malignant gynecologic neoplasms. Advanced staging and sarcomatous overgrowth predict poor prognosis. Because the genomic landscape remains poorly understood, we conducted this study to characterize the genomewide copy number variations in adenosarcomas. Sixteen tumors, including eight with and eight without sarcomatous overgrowth, were subjected to a molecular inversion probe array analysis. Copy number variations, particularly losses, were significantly higher in cases with sarcomatous overgrowth. Frequent gains of chromosomal 12q were noted, often involving cancer-associated genes CDK4 (six cases), MDM2, CPM, YEATS4, DDIT3, GLI1 (five each), HMGA2 and STAT6 (four), without association with sarcomatous overgrowth status. The most frequent losses involved chromosomes 13q (five cases), 9p, 16q and 17q (four cases each) and were almost limited to cases with sarcomatous overgrowth. MDM2 and CDK4 amplification, as well as losses of RB1 (observed in two cases) and CDKN2A/B (one case), was verified by FISH. By immunohistochemistry, all MDM2/CDK4-coamplified cases were confirmed to overexpress both encoded proteins, whereas all four cases with (plus an additional four without) gain of HMGA2 overexpressed the HMGA2 protein. Both cases with RB1 loss were negative for the immunostaining of the encoded protein. Chromothripsis-like copy number profiles involving chromosome 12 or 14 were observed in three fatal cases, all of which harbored sarcomatous overgrowth. With whole chromosome painting and deconvolution fluorescent microscopy, dividing tumor cells in all three cases were shown to have scattered extrachromosomal materials derived from chromosomes involved by chromothripsis, suggesting that this phenomenon may serve as visual evidence for chromothripsis in paraffin tissue. In conclusion, we identified frequent chromosome 12q amplifications, including loci containing potential pharmacological targets. Global chromosomal instability and chromothripsis were more frequent in cases with sarcomatous overgrowth. To our knowledge, this is the first time that evidence of chromothripsis has been demonstrated in paraffin-embedded clinical tissues and in adenosarcomas.
Collapse
Affiliation(s)
- Jen-Chieh Lee
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Tzu-Pin Lu
- Department of Public Health, Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun A Changou
- Integrated Laboratory, Center of Translational Medicine, Graduate Institute of Translational Medicine, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Cher-Wei Liang
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsien-Neng Huang
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Pathology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Alexandra Lauria
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hsuan-Ying Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chin-Yao Lin
- Department of Pathology, Cardinal Tien Hospital Yung-Ho Branch, New Taipei City, Taiwan
| | - Ying-Cheng Chiang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ben Davidson
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ming-Chieh Lin
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuan-Ting Kuo
- Department and Graduate Institute of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
21
|
Rebuzzini P, Zuccotti M, Redi CA, Garagna S. Achilles' heel of pluripotent stem cells: genetic, genomic and epigenetic variations during prolonged culture. Cell Mol Life Sci 2016; 73:2453-66. [PMID: 26961132 PMCID: PMC11108315 DOI: 10.1007/s00018-016-2171-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 01/28/2016] [Accepted: 02/25/2016] [Indexed: 12/12/2022]
Abstract
Pluripotent stem cells differentiate into almost any specialized adult cell type of an organism. PSCs can be derived either from the inner cell mass of a blastocyst-giving rise to embryonic stem cells-or after reprogramming of somatic terminally differentiated cells to obtain ES-like cells, named induced pluripotent stem cells. The potential use of these cells in the clinic, for investigating in vitro early embryonic development or for screening the effects of new drugs or xenobiotics, depends on capability to maintain their genome integrity during prolonged culture and differentiation. Both human and mouse PSCs are prone to genomic and (epi)genetic instability during in vitro culture, a feature that seriously limits their real potential use. Culture-induced variations of specific chromosomes or genes, are almost all unpredictable and, as a whole, differ among independent cell lines. They may arise at different culture passages, suggesting the absence of a safe passage number maintaining genome integrity and rendering the control of genomic stability mandatory since the very early culture passages. The present review highlights the urgency for further studies on the mechanisms involved in determining (epi)genetic and chromosome instability, exploiting the knowledge acquired earlier on other cell types.
Collapse
Affiliation(s)
- Paola Rebuzzini
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie 'Lazzaro Spallanzani', Università degli Studi di Pavia, Via Ferrata 9, 27100, Pavia, Italy.
- Center for Health Technologies (C.H.T.), Università degli Studi di Pavia, Via Ferrata 1, Pavia, Italy.
| | - Maurizio Zuccotti
- Unita' di Anatomia, Istologia ed Embriologia, Dipartimento di Scienze Biomediche, Biotecnologiche e Traslazionali (S.BI.BI.T.), Università degli Studi di Parma, Via Volturno 39, 43100, Parma, Italy.
| | - Carlo Alberto Redi
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie 'Lazzaro Spallanzani', Università degli Studi di Pavia, Via Ferrata 9, 27100, Pavia, Italy
- Center for Health Technologies (C.H.T.), Università degli Studi di Pavia, Via Ferrata 1, Pavia, Italy
- Fondazione I.R.C.C.S. Policlinico San Matteo, Piazzale Golgi, 19, 27100, Pavia, Italy
| | - Silvia Garagna
- Laboratorio di Biologia dello Sviluppo, Dipartimento di Biologia e Biotecnologie 'Lazzaro Spallanzani', Università degli Studi di Pavia, Via Ferrata 9, 27100, Pavia, Italy.
- Center for Health Technologies (C.H.T.), Università degli Studi di Pavia, Via Ferrata 1, Pavia, Italy.
| |
Collapse
|
22
|
David A, Amartely H, Rabinowicz N, Shamir M, Friedler A, Izraeli S. Molecular basis of the STIL coiled coil oligomerization explains its requirement for de-novo formation of centrosomes in mammalian cells. Sci Rep 2016; 6:24296. [PMID: 27075531 PMCID: PMC4830966 DOI: 10.1038/srep24296] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/24/2016] [Indexed: 11/09/2022] Open
Abstract
The STIL protein is essential for centriole replication and for the non-templated, de novo centriole biogenesis that is required for mammalian embryogenesis. Here we performed quantitative biophysical and structural analysis of the central short coiled coil domain (CCD) of STIL that is critical for its function. Using biophysical, biochemical and cell biology approaches, we identified the specific residues in the CCD that mediate the oligomerization, centrosomal localization and protein interactions of STIL. We characterized the structural properties of the coiled coil peptide using circular dichroism spectroscopy and size exclusion chromatography. We identified two regions in this domain, containing eight hydrophobic residues, which mediate the coiled coil oligomerization. Mutations in these residues destabilized the coiled coil thermodynamically but in most cases did not affect its secondary structure. Reconstituting mouse embryonic fibroblasts lacking endogenous Stil, we show that STIL oligomerization mediated by these residues is not only important for the centrosomal functions of STIL during the canonical duplication process but also for de-novo formation of centrosomes.
Collapse
Affiliation(s)
- Ahuvit David
- Sheba Cancer Research Center and the Edmond and Lily Safra Children Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel.,Department of molecular genetics and biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hadar Amartely
- Institute of Chemistry, the Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Noa Rabinowicz
- Sheba Cancer Research Center and the Edmond and Lily Safra Children Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel.,Department of molecular genetics and biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mai Shamir
- Institute of Chemistry, the Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Assaf Friedler
- Institute of Chemistry, the Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | - Shai Izraeli
- Sheba Cancer Research Center and the Edmond and Lily Safra Children Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel.,Department of molecular genetics and biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
23
|
Kryukova E, Kryukov F, Hajek R. Centrosome amplification and clonal evolution in multiple myeloma: Short review. Crit Rev Oncol Hematol 2015; 98:116-21. [PMID: 26589397 DOI: 10.1016/j.critrevonc.2015.10.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 09/14/2015] [Accepted: 10/28/2015] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is composed of an array of multiple clones, each potentially associated with different clinical behavior. Previous studies focused on clinical implication of centrosome amplification (CA) in MM show contradictory results. It seems that the role of CA as well as CA formation in MM differ from other malignancies. This has brought about a question about the role of CA positive clone which is--is it going to be a more aggressive clone evolutionally arising under pressure of negative conditions or can CA serve as a marker of cell abnormality and lead to cell death and further elimination of this damaged subpopulation? This current review is devoted to the discussion of the existence of MM subclones with centrosome amplification (CA), its evolutionary behaviour within intraclonal heterogeneity as well as its potential impact on the disease progression and MM treatment.
Collapse
Affiliation(s)
- Elena Kryukova
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, Czech Republic
| | - Fedor Kryukov
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, Czech Republic.
| | - Roman Hajek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava, Czech Republic
| |
Collapse
|
24
|
Upregulation of centromere protein H is associated with progression of renal cell carcinoma. J Mol Histol 2015; 46:377-85. [DOI: 10.1007/s10735-015-9635-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 08/03/2015] [Indexed: 12/27/2022]
|
25
|
Paulitschke V, Gerner C, Hofstätter E, Mohr T, Mayer RL, Pehamberger H, Kunstfeld R. Proteome profiling of keratinocytes transforming to malignancy. Electrophoresis 2015; 36:564-76. [PMID: 25395074 DOI: 10.1002/elps.201400309] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/20/2014] [Accepted: 10/29/2014] [Indexed: 01/19/2023]
Abstract
To shed light on the multistep process of squamous cell carcinoma development and the underlying pathologic mechanisms, we performed comparative proteome analysis of keratinocytes, keratinocytes stimulated with Il-1beta, and A431 epidermoid carcinoma cells. Fractionation of the cells into supernatant, nucleus, and cytoplasm was followed by protein separation, proteolytic digest, and nano-LC separation, and fragmentation using an ion trap mass spectrometer. Specific bioinformatics tools were used to generate a list of keratinocyte-specific proteins. Ninety percent of these proteins were found to be upregulated in keratinocytes versus the A431 cells. Classification of the identified proteins by biologic function and gene set enrichment analysis revealed that keratinocytes produced more proteins involved in cell differentiation, cell adhesion, cell junction, calcium ion, calmodulin binding, cytoskeleton organization, and cytokinesis, whereas A431 produced more proteins involved in cell cycle checkpoint, cell cycle process, RNA processing and transport, DNA damage and repair, RNA and DNA binding, and chromatin remodeling. The protein signatures of A431 and normal keratinocytes treated with IL-1beta showed marked similarity, confirming that inflammation is an important step in malignant transformation in nonmelanoma skin cancer. Thus, proteome profiling and bioinformatic processing may support the understanding of the underlying mechanisms, with the potential to facilitate development of early biomarkers and patient-tailored therapy.
Collapse
Affiliation(s)
- Verena Paulitschke
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
26
|
Huston RL. Using the Electromagnetics of Cancer’s Centrosome Clusters to Attract Therapeutic Nanoparticles. ACTA ACUST UNITED AC 2015. [DOI: 10.4236/abb.2015.63017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
27
|
Aghmesheh M, Saxena A, Niknam F. BRCA1 mutation site may be linked with nuclear DNA ploidy in BRCA1-mutated ovarian carcinomas. Asia Pac J Clin Oncol 2014; 11:135-41. [PMID: 25428229 DOI: 10.1111/ajco.12310] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2014] [Indexed: 11/29/2022]
Abstract
AIMS BRCA1 has a role in maintaining normal nuclear DNA content during cell division and its inactivation may result in DNA aneuploidy and cancer progression. BRCA1-linked breast cancers are more aneuploid and have a worse prognosis, but this has not been elucidated in ovarian cancers. This study explores the potential difference in ploidy status between BRCA1-mutated and sporadic ovarian carcinomas. It also explores the potential association between BRCA1 mutation site and DNA ploidy status. METHODS This study compared DNA ploidy status of tumor blocks from 23 BRCA1-mutated ovarian carcinomas with that of 23 sporadic ovarian carcinomas matched for histologic subtype, patient age, stage and grade. DNA content of the nuclei was measured by Feulgen-Schiff staining followed by image cytometry and compared. RESULTS BRCA1-linked tumors with a stop codon closer to the N-terminal (between 1 and 500 aa; 6/6, 100%) had a significantly higher frequency of nondiploidy compared with those with stop codon above 500 aa (7/12, 58%) (P = 0.033). A diploid peak was detected in 28% of BRCA1-mutated ovarian cancers and in 33% of sporadic ovarian cancers. CONCLUSIONS The present study concluded that ovarian tumors with mutations closer to the N-terminal of BRCA1 may have a higher risk of DNA aneuploidy. There is no significant difference between BRCA1-mutated and sporadic ovarian carcinomas with respect to the DNA content.
Collapse
Affiliation(s)
- Morteza Aghmesheh
- Illawarra Cancer Care Centre, Wollongong Hospital, Wollongong, New South Wales, Australia; Peter MacCallum Cancer Centre, Research Division, The Kathleen Cuningham Consortium for Research into Familial Breast Cancer, East Melbourne, Victoria, Australia
| | | | | |
Collapse
|
28
|
Douthwright S, Sluder G. Link between DNA damage and centriole disengagement/reduplication in untransformed human cells. J Cell Physiol 2014; 229:1427-36. [PMID: 24532022 PMCID: PMC4122266 DOI: 10.1002/jcp.24579] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 02/12/2014] [Indexed: 12/21/2022]
Abstract
The radiation and radiomimetic drugs used to treat human tumors damage DNA in both cancer cells and normal proliferating cells. Centrosome amplification after DNA damage is well established for transformed cell types but is sparsely reported and not fully understood in untransformed cells. We characterize centriole behavior after DNA damage in synchronized untransformed human cells. One hour treatment of S phase cells with the radiomimetic drug, Doxorubicin, prolongs G2 by at least 72 h, though 14% of the cells eventually go through mitosis in that time. By 72 h after DNA damage we observe a 52% incidence of centriole disengagement plus a 10% incidence of extra centrioles. We find that either APC/C or Plk activities can disengage centrioles after DNA damage, though they normally work in concert. All disengaged centrioles are associated with γ-tubulin and maturation markers and thus, should in principle be capable of reduplicating and organizing spindle poles. The low incidence of reduplication of disengaged centrioles during G2 is due to the p53-dependent expression of p21 and the consequent loss of Cdk2 activity. We find that 26% of the cells going through mitosis after DNA damage contain disengaged or extra centrioles. This could produce genomic instability through transient or persistent spindle multipolarity. Thus, for cancer patients the use of DNA damaging therapies raises the chances of genomic instability and evolution of transformed characteristics in proliferating normal cell populations.
Collapse
Affiliation(s)
- Stephen Douthwright
- Department of Cell and Developmental Biology University of Massachusetts Medical School, Worcester, Massachusetts 01655
| | - Greenfield Sluder
- Department of Cell and Developmental Biology University of Massachusetts Medical School, Worcester, Massachusetts 01655
| |
Collapse
|
29
|
Wang J, Xiao Z. Mesenchymal stem cells in pathogenesis of myelodysplastic syndromes. Stem Cell Investig 2014; 1:16. [PMID: 27358862 DOI: 10.3978/j.issn.2306-9759.2014.08.02] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 08/18/2014] [Indexed: 01/13/2023]
Abstract
Myelodysplastic syndromes (MDS) are clonal malignant stem cell disorders characterized by inefficient hematopoiesis. The role of the marrow microenvironment in the pathogenesis of the disease has been controversial. Emerging evidence indicated that mesenchymal stem cells (MSC) derived from MDS patients were cytogenetically abnormal, and they showed a deficient hematopoietic-supportive capacity and increased production of cytokine such as tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), interferon γ (IFN-γ). From the point of some evidence, the abnormal microenvironment seems to participate in the progression of the disease by contributing to the selective expansion of the malignant clone. In this review, we will discuss the most recent progress related to identification of normal MSC and the importance of the stem cell niche in development and maintenance of MDS.
Collapse
Affiliation(s)
- Jingya Wang
- 1 MDS and MPN Centre, 2 State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Zhijian Xiao
- 1 MDS and MPN Centre, 2 State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
30
|
Abstract
This paper describes the inner workings of centrioles (a pair of small organelles adjacent to the nucleus) as they create cell electropolarity, engage in cell division (mitosis), but in going awry, also promote the development of cancers. The electropolarity arises from vibrations of microtubules composing the centrioles. Mitosis begins as each centrioles duplicates itself by growing a daughter centriole on its side. If during duplication more than one daughter is grown, cancer can occur and the cells divide uncontrollably. Cancer cells with supernumerary centrioles have high electropolarity which can serve as an attractor for charged therapeutic nanoparticles.
Collapse
Affiliation(s)
- Ronald L. Huston
- Life Fellow ASME Department of Mechanical and Materials Engineering, University of Cincinnati, P.O. Box 210072, Cincinnati, OH 45221-0072 e-mail:
| |
Collapse
|
31
|
Zhao J, Zou Y, Liu H, Wang H, Zhang H, Hou W, Li X, Jia X, Zhang J, Hou L, Zhang B. TEIF associated centrosome activity is regulated by EGF/PI3K/Akt signaling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:1851-64. [PMID: 24769208 DOI: 10.1016/j.bbamcr.2014.04.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/29/2014] [Accepted: 04/17/2014] [Indexed: 10/25/2022]
Abstract
Centrosome amplification, which is a characteristic of cancer cells, has been understood as a driving force of genetic instability in the development of cancer. In previous work, we demonstrated that TEIF (transcriptional element-interacting factor) distributes in the centrosomes and regulates centrosome status under both physiologic and pathologic conditions. Here we identify TEIF as a downstream effector in EGF/PI3K/Akt signaling. The addition of EGF or transfection of active Akt stimulates centrosome TEIF distribution, resulting in an increase of centrosome splitting and amplification, while inhibitors of either PI3K or Akt attenuate these changes in TEIF and the associated centrosome status. A consensus motif for Akt phosphorylation (RHRVLT) proved to be involved in centrosomal TEIF localization, and the 469-threonine of this motif may be phosphorylated by Akt both in vitro and in vivo. Elimination of this phosphorylated site on TEIF caused reduced centrosome distribution and centrosome splitting or amplification. Moreover, TEIF closely co-localized with C-NAP1 at the proximal ends of centrioles, and centriolar loading of TEIF stimulated by EGF/Akt could displace C-NAP1, resulting in centrosome splitting. These findings reveal linkage of the EGF/PI3K/Akt signaling pathway to regulation of centrosome status which may act as an oncogenic pathway and induce genetic instability in carcinogenesis.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Yongxin Zou
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Haijing Liu
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Huali Wang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Hong Zhang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Wei Hou
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Xin Li
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Xinying Jia
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Jing Zhang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Lin Hou
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Bo Zhang
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Haidian District, Beijing 100191, China.
| |
Collapse
|
32
|
Intratumoral Hypoxia as the Genesis of Genetic Instability and Clinical Prognosis in Prostate Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 772:189-204. [DOI: 10.1007/978-1-4614-5915-6_9] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
33
|
Chopra A, Anderson A, Giardina C. Novel piperazine-based compounds inhibit microtubule dynamics and sensitize colon cancer cells to tumor necrosis factor-induced apoptosis. J Biol Chem 2013; 289:2978-91. [PMID: 24338023 DOI: 10.1074/jbc.m113.499319] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
We recently identified a series of mitotically acting piperazine-based compounds that potently increase the sensitivity of colon cancer cells to apoptotic ligands. Here we describe a structure-activity relationship study on this compound class and identify a highly active derivative ((4-(3-chlorophenyl)piperazin-1-yl)(2-ethoxyphenyl)methanone), referred to as AK301, the activity of which is governed by the positioning of functional groups on the phenyl and benzoyl rings. AK301 induced mitotic arrest in HT29 human colon cancer cells with an ED50 of ≈115 nm. Although AK301 inhibited growth of normal lung fibroblast cells, mitotic arrest was more pronounced in the colon cancer cells (50% versus 10%). Cells arrested by AK301 showed the formation of multiple microtubule organizing centers with Aurora kinase A and γ-tubulin. Employing in vitro and in vivo assays, tubulin polymerization was found to be slowed (but not abolished) by AK301. In silico molecular docking suggests that AK301 binds to the colchicine-binding domain on β-tubulin, but in a novel orientation. Cells arrested by AK301 expressed elevated levels of TNFR1 on their surface and more readily activated caspases-8, -9, and -3 in the presence of TNF. Relative to other microtubule destabilizers, AK301 was the most active TNF-sensitizing agent and also stimulated Fas- and TRAIL-induced apoptosis. In summary, we report a new class of mitosis-targeting agents that effectively sensitizes cancer cells to apoptotic ligands. These compounds should help illuminate the role of microtubules in regulating apoptotic ligand sensitivity and may ultimately be useful for developing agents that augment the anti-cancer activities of the immune response.
Collapse
|
34
|
Lu G, Shan T, He S, Ren M, Zhu M, Hu Y, Lu X, Zhang D. Overexpression of CENP-H as a novel prognostic biomarker for human hepatocellular carcinoma progression and patient survival. Oncol Rep 2013; 30:2238-44. [PMID: 23970101 DOI: 10.3892/or.2013.2675] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Accepted: 07/09/2013] [Indexed: 11/06/2022] Open
Abstract
Centromere protein H (CENP-H) has been shown to be significantly upregulated in many types of cancers and is associated with disrupted cell cycle regulation, cell proliferation and genetic instability. The aim of the present study was to explore the expression and localization of CENP-H in hepatocellular carcinoma (HCC) and determine whether its overexpression is a prognostic biomarker for HCC. Reverse transcription-polymerase chain reaction (pcr), real-time qPCR and western blotting were used to compare CENP-H expression at the mRNA and protein levels in HCC samples and corresponding adjacent non-cancerous samples. CENP-H protein levels were determined in 60 paired paraffin-embedded HCC tissues using immunohistochemistry (IHC), and the correlation with clinicopathological features and patient prognosis was analyzed. In addition, an immunofluorescence assay was performed to test the expression and localization of CENP-H protein in HCC cells. Results showed that levels of CENP-H mRNA and protein were higher in HCC samples than in the corresponding adjacent non-cancerous samples. In 60 paired paraffin-embedded tissues, CENP-H was upregulated in the HCC samples (38/60, 63.3%) relative to the adjacent non-cancerous samples (21/60, 35%, P=0.003), and a higher level of upregulation was associated with tumor size (P=0.032); higher histological grade (P=0.001); more advanced TNM stage (P=0.002) and Chinese clinical stage (P=0.008); and poorer prognosis. In addition, consistent with the results of IHC, the immunofluorescence assay showed that CENP-H was localized in the nucleus of Hep3B cells. CENP-H was overexpressed in HCC, and its level of upregulation was an independent prognostic indicator, suggesting that CENP-H may be an effective therapeutic strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Guifang Lu
- Department of Gastroenterology, First Affiliated Hospital of the Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Zhang P, Ma X, Song E, Chen W, Pang H, Ni D, Gao Y, Fan Y, Ding Q, Zhang Y, Zhang X. Tubulin cofactor A functions as a novel positive regulator of ccRCC progression, invasion and metastasis. Int J Cancer 2013; 133:2801-11. [PMID: 23740643 DOI: 10.1002/ijc.28306] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 05/10/2013] [Indexed: 11/11/2022]
Abstract
Microtubules (Mts), which consist of α/β-tubulin heterodimers, are involved in cancer development and metastasis. Tubulin cofactor A (TBCA) plays crucial roles in modulating tubulin folding and α/β-tubulin heterodimer polymerization. Here, we identified the aberrant expression of TBCA in clear cell renal cell carcinoma (ccRCC) specimens as well as cell lines and revealed the function of TBCA as a novel positive regulator in ccRCC progression, invasion and metastasis. qRT-PCR, Western blot and immunohistochemistry assays confirmed that TBCA was significantly highly expressed in ccRCC specimens and cell lines compared to their corresponding normal kidney tissues and HKC. Accordingly, the influence of TBCA on cell proliferation, apoptosis and invasion/migration was detected through overexpression and knockdown of endogenous TBCA protein level in ccRCC cells via plasmids. Silencing of TBCA expression inhibited the proliferation of 786-O cells and Caki-1 cells and promoted the apoptosis of 786-O cells. Down-regulation of TBCA expression also reduced the invasion and migration ability of 786-O cells. Interestingly, overexpression of TBCA did not induce biocharacteristics that directly contrasted to those of TBCA knockdown. Importantly, exploration of the mechanism showed that TBCA could function via modulating cytoskeleton integration and influencing cell cycle progress. Furthermore, down-regulation of TBCA expression in 786-O and Caki-1 cells affected cytoskeleton integration and cell size, induced S/G2 cell cycle arrest and led to cyclineA/E and CDK2 aberrant expression. By investigating novel roles of TBCA in regulation of ccRCC cell progression, invasion and metastasis, our study identified that TBCA may be a potential molecular target for ccRCC therapy.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Urology, PLA General Hospital, Beijing, People's Republic of China; State Key Laboratory of Kidney Diseases, PLA General Hospital, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ha GH, Kim JL, Breuer EKY. Transforming acidic coiled-coil proteins (TACCs) in human cancer. Cancer Lett 2013; 336:24-33. [PMID: 23624299 DOI: 10.1016/j.canlet.2013.04.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 04/11/2013] [Accepted: 04/16/2013] [Indexed: 10/26/2022]
Abstract
Fine-tuned regulation of the centrosome/microtubule dynamics during mitosis is essential for faithful cell division. Thus, it is not surprising that deregulations in this dynamic network can contribute to genomic instability and tumorigenesis. Indeed, centrosome loss or amplification, spindle multipolarity and aneuploidy are often found in a majority of human malignancies, suggesting that defects in centrosome and associated microtubules may be directly or indirectly linked to cancer. Therefore, future research to identify and characterize genes required for the normal centrosome function and microtubule dynamics may help us gain insight into the complexity of cancer, and further provide new avenues for prognostic, diagnostics and therapeutic interventions. Members of the transforming acidic coiled-coil proteins (TACCs) family are emerging as important players of centrosome and microtubule-associated functions. Growing evidence indicates that TACCs are involved in the progression of certain solid tumors. Here, we will discuss our current understanding of the biological function of TACCs, their relevance to human cancer and possible implications for cancer management.
Collapse
Affiliation(s)
- Geun-Hyoung Ha
- Oncology Institute, Cardinal Bernardin Cancer Center, Loyola University Medical Center, Maywood, IL 60153, USA
| | | | | |
Collapse
|
37
|
Maier B, Kirsch M, Anderhub S, Zentgraf H, Krämer A. The novel actin/focal adhesion-associated protein MISP is involved in mitotic spindle positioning in human cells. Cell Cycle 2013; 12:1457-71. [PMID: 23574715 PMCID: PMC3674073 DOI: 10.4161/cc.24602] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Accurate mitotic spindle positioning is essential for the regulation of cell fate choices, cell size and cell position within tissues. The most prominent model of spindle positioning involves a cortical pulling mechanism, where the minus end-directed microtubule motor protein dynein is attached to the cell cortex and exerts pulling forces on the plus ends of astral microtubules that reach the cortex. In nonpolarized cultured cells integrin-dependent, retraction fiber-mediated cell adhesion is involved in spindle orientation. Proteins serving as intermediaries between cortical actin or retraction fibers and astral microtubules remain largely unknown. In a recent genome-wide RNAi screen we identified a previously uncharacterized protein, MISP (C19ORF21) as being involved in centrosome clustering, a process leading to the clustering of supernumerary centrosomes in cancer cells into a bipolar mitotic spindle array by microtubule tension. Here, we show that MISP is associated with the actin cytoskeleton and focal adhesions and is expressed only in adherent cell types. During mitosis MISP is phosphorylated by Cdk1 and localizes to retraction fibers. MISP interacts with the +TIP EB1 and p150glued, a subunit of the dynein/dynactin complex. Depletion of MISP causes mitotic arrest with reduced tension across sister kinetochores, chromosome misalignment and spindle multipolarity in cancer cells with supernumerary centrosomes. Analysis of spindle orientation revealed that MISP depletion causes randomization of mitotic spindle positioning relative to cell axes and cell center. Together, we propose that MISP links microtubules to the actin cytoskeleton and focal adhesions in order to properly position the mitotic spindle.
Collapse
Affiliation(s)
- Bettina Maier
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
38
|
Brodin BA. CINSARC: a new look into an old concept gives hope for new treatments for synovial sarcomas. Transl Pediatr 2013; 2:70-2. [PMID: 26835293 PMCID: PMC4728922 DOI: 10.3978/j.issn.2224-4336.2013.04.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Bertha A Brodin
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm 171 76, Sweden
| |
Collapse
|
39
|
Clinical implication of centrosome amplification and expression of centrosomal functional genes in multiple myeloma. J Transl Med 2013; 11:77. [PMID: 23522059 PMCID: PMC3615957 DOI: 10.1186/1479-5876-11-77] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 03/10/2013] [Indexed: 12/04/2022] Open
Abstract
Background Multiple myeloma (MM) is a low proliferative tumor of postgerminal center plasma cell (PC). Centrosome amplification (CA) is supposed to be one of the mechanisms leading to chromosomal instability. Also, CA is associated with deregulation of cell cycle, mitosis, DNA repair and proliferation. The aim of our study was to evaluate the prognostic significance and possible role of CA in pathogenesis and analysis of mitotic genes as mitotic disruption markers. Design and methods A total of 173 patients were evaluated for this study. CD138+ cells were separated by MACS. Immunofluorescent labeling of centrin was used for evaluation of centrosome amplification in PCs. Interphase FISH with cytoplasmic immunoglobulin light chain staining (cIg FISH) and qRT-PCR were performed on PCs. Results Based on the immunofluorescent staining results, all patients were divided into two groups: CA positive (38.2%) and CA negative (61.8%). Among the newly diagnosed patients, worse overall survival was indicated in the CA negative group (44/74) in comparison to the CA positive group (30/74) (P = 0.019). Gene expression was significantly down-regulated in the CA positive group in comparison to CA negative in the following genes: AURKB, PLK4, TUBG1 (P < 0.05). Gene expression was significantly down-regulated in newly diagnosed in comparison to relapsed patients in the following genes: AURKA, AURKB, CCNB1, CCNB2, CETN2, HMMR, PLK4, PCNT, and TACC3 (P < 0.05). Conclusions Our findings indicate better prognosis for CA positive newly diagnosed patients. Considering revealed clinical and gene expression heterogeneity between CA negative and CA positive patients, there is a possibility to characterize centrosome amplification as a notable event in multiple myeloma pathogenesis.
Collapse
|
40
|
|
41
|
Kryukov F, Nemec P, Dementyeva E, Kubiczkova L, Ihnatova I, Budinska E, Jarkovsky J, Sevcikova S, Kuglik P, Hajek R. Molecular heterogeneity and centrosome-associated genes in multiple myeloma. Leuk Lymphoma 2013; 54:1982-8. [DOI: 10.3109/10428194.2013.764416] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
42
|
Zhang KJ, Wang M. Potential effects of CRM1 inhibition in mantle cell lymphoma. Chin J Cancer Res 2013; 24:374-87. [PMID: 23357869 DOI: 10.3978/j.issn.1000-9604.2012.09.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 02/08/2012] [Indexed: 12/14/2022] Open
Abstract
Mantle cell lymphoma (MCL) is an aggressive histotype of B-cell non-Hodgkin lymphoma. The disease has no known cure, which prompts the urgent need for novel therapeutic agents. Chromosomal region maintenance 1 (CRM1) may play a role in human neoplasia and serve as a novel target of cancer treatment. This study summarizes MCL pathogenesis and determines the involvement of CRM1 in the regulation of several vital signaling pathways contributing to MCL pathogenesis, including the pathways of cell cycle progression, DNA damage response, phosphoinositide kinase-3, nuclear factor-κB activation, and chromosomal stability. A preclinical study is also presented to compare the CRM1 status in MCL cell lines and primary MCL cells with normal B cells, as well as the therapeutic efficiency of CRM1 inhibition in MCL in vitro and in vivo, which make these agents potential targets of novel MCL treatments.
Collapse
Affiliation(s)
- Ke-Jie Zhang
- Department of Hematology, Zhongshan Hospital, Xiamen University, Xiamen 361004, China; ; Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston 77030, USA
| | | |
Collapse
|
43
|
Bogen KT. Efficient tumorigenesis by mutation-induced failure to terminate microRNA-mediated adaptive hyperplasia. Med Hypotheses 2012. [PMID: 23183421 DOI: 10.1016/j.mehy.2012.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Seven current contending cancer theories consider different sets of critical events as sufficient for tumorigenesis. These theories, most recently the microRNA dysregulation (MRD) theory, have overlapping attributes and extensive empirical support, but also some discrepancies, and some do not address both benign and malignant tumorigenesis. By definition, the most efficient tumorigenic pathways will dominate under conditions that selectively activate those pathways. The MRD theory provides a mechanistic basis to combine elements of the current theories into a new hypothesis that: (i) tumors arise most efficiently under stress that induces and sustains either protective or regenerative states of adaptive hyperplasia (AH) that normally are epigenetically maintained unless terminated; and (ii) if dysregulated by a somatic mutation that prevents normal termination, these two AH states can generate benign and malignant tumors, respectively. This hypothesis, but not multistage cancer theory, predicts that key participating AH-stem-cell populations expand markedly when triggered by stress, particularly chronic metabolic or oxidative stress, mechanical irritation, toxic exposure, wounding, inflammation, and/or infection. This hypothesis predicts that microRNA expression patterns in benign vs. malignant tumor tissue will correlate best with those governing protective vs. regenerative AH in that tissue, and that tumors arise most efficiently inmutagen-exposed stem cells that either happen to be in, or incidentally later become recruited into, an AH state.
Collapse
Affiliation(s)
- Kenneth T Bogen
- DrPH DABT, Exponent Inc., Health Sciences, 475, 14th Street, Ste 400, Oakland, CA 94612, USA.
| |
Collapse
|
44
|
Zhu Y, Wang C, Lan J, Yu J, Jin C, Huang H. Phosphorylation of Tara by Plk1 is essential for faithful chromosome segregation in mitosis. Exp Cell Res 2012; 318:2344-52. [PMID: 22820163 DOI: 10.1016/j.yexcr.2012.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 06/27/2012] [Accepted: 07/04/2012] [Indexed: 11/16/2022]
Abstract
Trio-associated repeat on actin (Tara) is an F-actin binding protein and regulates actin cytoskeletal organization. In our previous study, we have found that Tara associates with telomeric repeat binding factor 1 (TRF1) and mediates the function of TRF1 in mitotic regulation. We also found that overexpression HECTD3, a member of HECT E3 ubiquitin ligases, enhances the ubiquitination of Tara in vivo and promotes the degradation of Tara, and such degradation of Tara facilitates cell cycle progression. However, less is known about the post-translational modification of Tara in mitosis. Here we show that Tara is a novel Polo-like kinase 1 (Plk1) target protein. Plk1 interacts with and phosphorylates Tara in vivo and in vitro. Actually, the Thr-457 in Tara was a bona fide in vivo phosphorylation site for Plk1. Interestingly, we found that the centrosomal localization of Tara depended on the Thr-457 phosphorylation and the kinase activity of Plk1. Furthermore, overexpression of non-phosphorylatable mutant of Tara caused aberrant mitosis delay in HeLa cells. Our study demonstrated that Plk1-mediated phospho-dependent centrosomal localization of Tara is important for faithful chromosome segregation, and provided novel insights into understanding on the role of Plk1 in cooperation with Tara in mitotic progression.
Collapse
Affiliation(s)
- Yuanyuan Zhu
- The First Affiliated Hospital of Zhejiang University Medical School, Hangzhou 310003, China
| | | | | | | | | | | |
Collapse
|
45
|
Raab MS, Breitkreutz I, Anderhub S, Rønnest MH, Leber B, Larsen TO, Weiz L, Konotop G, Hayden PJ, Podar K, Fruehauf J, Nissen F, Mier W, Haberkorn U, Ho AD, Goldschmidt H, Anderson KC, Clausen MH, Krämer A. GF-15, a novel inhibitor of centrosomal clustering, suppresses tumor cell growth in vitro and in vivo. Cancer Res 2012; 72:5374-85. [PMID: 22942257 DOI: 10.1158/0008-5472.can-12-2026] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In contrast to normal cells, malignant cells are frequently aneuploid and contain multiple centrosomes. To allow for bipolar mitotic division, supernumerary centrosomes are clustered into two functional spindle poles in many cancer cells. Recently, we have shown that griseofulvin forces tumor cells with supernumerary centrosomes to undergo multipolar mitoses resulting in apoptotic cell death. Here, we describe the characterization of the novel small molecule GF-15, a derivative of griseofulvin, as a potent inhibitor of centrosomal clustering in malignant cells. At concentrations where GF-15 had no significant impact on tubulin polymerization, spindle tension was markedly reduced in mitotic cells upon exposure to GF-15. Moreover, isogenic cells with conditional centrosome amplification were more sensitive to GF-15 than parental controls. In a wide array of tumor cell lines, mean inhibitory concentrations (IC(50)) for proliferation and survival were in the range of 1 to 5 μmol/L and were associated with apoptotic cell death. Importantly, treatment of mouse xenograft models of human colon cancer and multiple myeloma resulted in tumor growth inhibition and significantly prolonged survival. These results show the in vitro and in vivo antitumor efficacy of a prototype small molecule inhibitor of centrosomal clustering and strongly support the further evaluation of this new class of molecules.
Collapse
Affiliation(s)
- Marc S Raab
- Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Vulprecht J, David A, Tibelius A, Castiel A, Konotop G, Liu F, Bestvater F, Raab MS, Zentgraf H, Izraeli S, Krämer A. STIL is required for centriole duplication in human cells. J Cell Sci 2012; 125:1353-62. [PMID: 22349705 DOI: 10.1242/jcs.104109] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Centrioles are key structural elements of centrosomes and primary cilia. In mammals, only a few proteins including PLK4, CPAP (CENPJ), SAS6, CEP192, CEP152 and CEP135 have thus far been identified to be required for centriole duplication. STIL (SCL/TAL1 interrupting locus, also known as SIL) is a centrosomal protein that is essential for mouse and zebrafish embryonic development and mutated in primary microcephaly. Here, we show that STIL localizes to the pericentriolar material surrounding parental centrioles. Its overexpression results in excess centriole formation. siRNA-mediated depletion of STIL leads to loss of centrioles and abrogates PLK4-induced centriole overduplication. Additionally, we show that STIL is necessary for SAS6 recruitment to centrioles, suggesting that it is essential for daughter centriole formation, interacts with the centromere protein CPAP and rapidly shuttles between the cytoplasm and centrioles. Consistent with the requirement of centrioles for cilia formation, Stil(-/-) mouse embryonic fibroblasts lack primary cilia--a phenotype that can be reverted by restoration of STIL expression. These findings demonstrate that STIL is an essential component of the centriole replication machinery in mammalian cells.
Collapse
Affiliation(s)
- Julia Vulprecht
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Dept. of Internal Medicine V, University of Heidelberg, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
CHEN RONGMING, SHIH KUEICHUNG, HU ROUHMEI, TSAI JEFFREYJP. PREDICTING CANCEROUS GENES BASED ON REGULATION TRUTH TABLES. INT J ARTIF INTELL T 2011. [DOI: 10.1142/s0218213006002928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Several of ten thousands functional genes control the growth, genetics, and behavior of living organisms by regulating different gene expressions. The genes in a normal cell control the process of cell growth, differentiation, reproduction, and apoptosis via multiple steps of interactive regulation mechanism. The mechanism of gene regulation is a very important process in human beings. If there is something wrong in the gene regulation mechanism, it may cause some diseases such as cancer. It is very difficult to identify the regulatory relations among genes in human genome. Traditional biological research methods consume huge amount of time and man strength to do this work. In recent years, with the rapid development of microarray technologies, cDNA can be used to analyze the changes of gene expressions in different cells in a high throughput manner. In this paper, we propose a novel bioinformatics approach to predict possible cancerous genes based on a so-called regulation truth table (RTT) of genes. The RTT of two genes is constructed using the differential expressions of cDNA microarray data for tumor and normal tissues. The differences in regulatory relations of genes for tumor and normal tissues are adopted to identify possible cancerous genes.
Collapse
Affiliation(s)
- RONG-MING CHEN
- Department of Computer Science and Information Engineering, National University of Tainan, Tainan, Taiwan 70005, ROC
| | - KUEI-CHUNG SHIH
- Department of Computer Science, National Tsing Hua University, Hsinchu, Taiwan 30013, ROC
| | - ROUH-MEI HU
- Department of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan 41354, ROC
| | - JEFFREY J. P. TSAI
- Department of Computer Science, University of Illinois, Chicago, IL 60607, USA
| |
Collapse
|
48
|
Krzywicka-Racka A, Sluder G. Repeated cleavage failure does not establish centrosome amplification in untransformed human cells. ACTA ACUST UNITED AC 2011; 194:199-207. [PMID: 21788368 PMCID: PMC3144409 DOI: 10.1083/jcb.201101073] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Transient cleavage failure in dividing cells is not sufficient to establish stable populations of cells with extra centrosomes. We tested whether cleavage failure as a transient event establishes an incidence of centrosome amplification in cell populations. Five rounds of ∼30% cytochalasin-induced cleavage failure in untransformed human cell cultures did not establish centrosome amplification in the short or long terms. The progeny of binucleate cells progressively dropped out of the cell cycle and expressed p53/p21, and none divided a fourth time. We also tested whether cleavage failure established centrosome amplification in transformed cell populations. Tetraploid HCT116 p53−/− cells eventually all failed cleavage repeatedly and ceased proliferating. HeLa cells all died or arrested within four cell cycles. Chinese hamster ovary cells proliferated after cleavage failure, but five rounds of induced cleavage failure produced a modest increase in the incidence of centrosome amplification in the short term, which did not rise with more cycles of cleavage failure. This incidence dropped to close to control values in the long term despite a 2–6% rate of spontaneous cleavage failure in the progeny of tetraploid cells.
Collapse
Affiliation(s)
- Anna Krzywicka-Racka
- Department of Cell Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | |
Collapse
|
49
|
Gualtieri M, Ovrevik J, Mollerup S, Asare N, Longhin E, Dahlman HJ, Camatini M, Holme JA. Airborne urban particles (Milan winter-PM2.5) cause mitotic arrest and cell death: Effects on DNA, mitochondria, AhR binding and spindle organization. Mutat Res 2011; 713:18-31. [PMID: 21645525 DOI: 10.1016/j.mrfmmm.2011.05.011] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Revised: 05/13/2011] [Accepted: 05/20/2011] [Indexed: 05/22/2023]
Abstract
Airborne particulate matter (PM) is considered to be an important contributor to lung diseases. In the present study we report that Milan winter-PM2.5 inhibited proliferation in human bronchial epithelial cells (BEAS-2B) by inducing mitotic arrest. The cell cycle arrest was followed by an increase in mitotic-apoptotic cells, mitotic slippage and finally an increase in "classical" apoptotic cells. Exposure to winter-PM10 induced only a slight effect which may be due to the presence of PM2.5 in this fraction while pure combustion particles failed to disturb mitosis. Fewer cells expressing the mitosis marker phospho-histone H3 compared to cells with condensed chromosomes, suggest that PM2.5 induced premature mitosis. PM2.5 was internalized into the cells and often localized in laminar organelles, although particles without apparent plasma membrane covering were also seen. In PM-containing cells mitochondria and lysosomes were often damaged, and in mitotic cells fragmented chromosomes often appeared. PM2.5 induced DNA strands breaks and triggered a DNA-damage response characterized by increased phosphorylation of ATM, Chk2 and H2AX; as well as induced a marked increase in expression of the aryl hydrocarbon receptor (AhR)-regulated genes, CYP1A1, CYP1B1 and AhRR. Furthermore, some disturbance of the organization of microtubules was indicated. It is hypothesized that the induced mitotic arrest and following cell death was due to a premature chromosome condensation caused by a combination of DNA, mitochondrial and spindle damage.
Collapse
Affiliation(s)
- Maurizio Gualtieri
- Applied Cell Biology and Particles Effects, Department of Environmental Science, University Milano-Bicocca, Piazza della Scienza 1, 20126 Milano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kryukov F, Dementyeva E, Smetana J, Sevcikova S, Kuglik P, Hajek R. Association of aneuploidy category with centrosome amplification in multiple myeloma. Leuk Lymphoma 2011; 52:2020-2. [PMID: 21635202 DOI: 10.3109/10428194.2011.587561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|