1
|
Thangaretnam K, Islam MO, Lv J, El-Rifai A, Perloff A, Soutto HL, Peng D, Chen Z. WEE1 inhibition in cancer therapy: Mechanisms, synergies, preclinical insights, and clinical trials. Crit Rev Oncol Hematol 2025; 211:104710. [PMID: 40187712 DOI: 10.1016/j.critrevonc.2025.104710] [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: 03/04/2025] [Accepted: 03/22/2025] [Indexed: 04/07/2025] Open
Abstract
WEE1 is a serine/threonine kinase that regulates the G2/M checkpoint by phosphorylating CDK1, preventing premature mitotic entry and maintaining genomic stability. Many cancers, particularly those with TP53 mutations, upregulate WEE1 to counteract replication stress and DNA damage, making it a key target for therapy. WEE1 inhibitors, especially adavosertib (AZD1775), have shown strong preclinical and clinical activity in ovarian, breast, gastrointestinal, and head and neck cancers. By inducing mitotic catastrophe and increasing DNA damage, WEE1 inhibition enhances the effectiveness of chemotherapies, including platinum-based agents, antimetabolites, and PARP inhibitors. It also synergizes with radiotherapy and immune checkpoint inhibitors, improving responses in tumors with immune evasion. However, challenges such as acquired resistance, toxicity, and patient selection remain obstacles to clinical implementation. Given the expanding role of WEE1 inhibitors in cancer treatment, a comprehensive review is needed to summarize their biological functions, structural regulation, and therapeutic applications. This review highlights key findings from preclinical and clinical studies, explores emerging biomarkers for patient stratification, and discusses strategies to overcome resistance and toxicity. By integrating current knowledge, we aim to provide insights into optimizing WEE1-targeted therapies and guiding future research to maximize their clinical impact in cancer treatment.
Collapse
Affiliation(s)
- Krishnapriya Thangaretnam
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Md Obaidul Islam
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Jialun Lv
- Gastric Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | | | - Ava Perloff
- College of Arts and Sciences, University of Miami, FL 33136, USA
| | - Houda L Soutto
- College of Arts and Sciences, University of Miami, FL 33136, USA
| | - Dunfa Peng
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Zheng Chen
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA.
| |
Collapse
|
2
|
Curran S, Dey G, Rees P, Nurse P. A quantitative and spatial analysis of cell cycle regulators during the fission yeast cycle. Proc Natl Acad Sci U S A 2022; 119:e2206172119. [PMID: 36037351 PMCID: PMC9457408 DOI: 10.1073/pnas.2206172119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/27/2022] [Indexed: 11/23/2022] Open
Abstract
We have carried out a systems-level analysis of the spatial and temporal dynamics of cell cycle regulators in the fission yeast Schizosaccharomyces pombe. In a comprehensive single-cell analysis, we have precisely quantified the levels of 38 proteins previously identified as regulators of the G2 to mitosis transition and of 7 proteins acting at the G1- to S-phase transition. Only 2 of the 38 mitotic regulators exhibit changes in concentration at the whole-cell level: the mitotic B-type cyclin Cdc13, which accumulates continually throughout the cell cycle, and the regulatory phosphatase Cdc25, which exhibits a complex cell cycle pattern. Both proteins show similar patterns of change within the nucleus as in the whole cell but at higher concentrations. In addition, the concentrations of the major fission yeast cyclin-dependent kinase (CDK) Cdc2, the CDK regulator Suc1, and the inhibitory kinase Wee1 also increase in the nucleus, peaking at mitotic onset, but are constant in the whole cell. The significant increase in concentration with size for Cdc13 supports the view that mitotic B-type cyclin accumulation could act as a cell size sensor. We propose a two-step process for the control of mitosis. First, Cdc13 accumulates in a size-dependent manner, which drives increasing CDK activity. Second, from mid-G2, the increasing nuclear accumulation of Cdc25 and the counteracting Wee1 introduce a bistability switch that results in a rapid rise of CDK activity at the end of G2 and thus, brings about an orderly progression into mitosis.
Collapse
Affiliation(s)
- Scott Curran
- Cell Cycle Laboratory, The Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - Gautam Dey
- Medical Research Council Laboratory for Molecular Cell Biology, London, WC1E 6BT, United Kingdom
| | - Paul Rees
- College of Engineering, Swansea University, Swansea, SA1 8EN, United Kingdom
- Imaging Platform Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Paul Nurse
- Cell Cycle Laboratory, The Francis Crick Institute, London, NW1 1AT, United Kingdom
- Laboratory of Yeast Genetics and Cell Biology, Rockefeller University, New York, NY 10065
| |
Collapse
|
3
|
Abstract
The centrosome is a multifunctional organelle that is known primarily for its microtubule organising function. Centrosomal defects caused by changes in centrosomal structure or number have been associated with human diseases ranging from congenital defects to cancer. We are only beginning to appreciate how the non-microtubule organising roles of the centrosome are related to these clinical conditions. In this review, we will discuss the historical evidence that led to the proposal that the centrosome participates in cell cycle regulation. We then summarize the body of work that describes the involvement of the mammalian centrosome in triggering cell cycle progression and checkpoint signalling. Then we will highlight work from the fission yeast model organism, revealing the molecular details that explain how the spindle pole body (SPB, the yeast functional equivalent of the centrosome), participates in these cell cycle transitions. Importantly, we will discuss some of the emerging questions from recent discoveries related to the role of the centrosome as a cell cycle regulator.
Collapse
|
4
|
Aguilar-Morante D, Gómez-Cabello D, Quek H, Liu T, Hamerlik P, Lim YC. Therapeutic Opportunities of Disrupting Genome Integrity in Adult Diffuse Glioma. Biomedicines 2022; 10:biomedicines10020332. [PMID: 35203541 PMCID: PMC8869545 DOI: 10.3390/biomedicines10020332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/28/2022] [Accepted: 01/28/2022] [Indexed: 11/09/2022] Open
Abstract
Adult diffuse glioma, particularly glioblastoma (GBM), is a devastating tumor of the central nervous system. The existential threat of this disease requires on-going treatment to counteract tumor progression. The present outcome is discouraging as most patients will succumb to this disease. The low cure rate is consistent with the failure of first-line therapy, radiation and temozolomide (TMZ). Even with their therapeutic mechanism of action to incur lethal DNA lesions, tumor growth remains undeterred. Delivering additional treatments only delays the inescapable development of therapeutic tolerance and disease recurrence. The urgency of establishing lifelong tumor control needs to be re-examined with a greater focus on eliminating resistance. Early genomic and transcriptome studies suggest each tumor subtype possesses a unique molecular network to safeguard genome integrity. Subsequent seminal work on post-therapy tumor progression sheds light on the involvement of DNA repair as the causative contributor for hypermutation and therapeutic failure. In this review, we will provide an overview of known molecular factors that influence the engagement of different DNA repair pathways, including targetable vulnerabilities, which can be exploited for clinical benefit with the use of specific inhibitors.
Collapse
Affiliation(s)
- Diana Aguilar-Morante
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain; (D.A.-M.); (D.G.-C.)
| | - Daniel Gómez-Cabello
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain; (D.A.-M.); (D.G.-C.)
| | - Hazel Quek
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia;
| | - Tianqing Liu
- NICM Health Research Institute, Westmead, NSW 2145, Australia;
| | | | - Yi Chieh Lim
- Danish Cancer Society, 2100 København, Denmark;
- Correspondence: ; Tel.: +45-35-257-413
| |
Collapse
|
5
|
Li J, Chen B, Xi WQ, Yang C, Zhang WX. Gene-Gene Interactions of Gemcitabine Metabolizing-Enzyme Genes hCNT3 and WEE1 for Preventing Severe Gemcitabine-Induced Hematological Toxicity. J Clin Pharmacol 2021; 61:1376-1385. [PMID: 33974709 DOI: 10.1002/jcph.1909] [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: 02/10/2021] [Accepted: 05/10/2021] [Indexed: 11/12/2022]
Abstract
Most patients experience severe hematological toxicity during treatment with gemcitabine; thus, preventing such toxicity would improve the treatment effects and patient quality of life. We analyzed 13 polymorphisms in the transporters, metabolizing enzymes, targets, and genes involved in DNA damage and the folate pathway among 132 patients treated with gemcitabine and studied their association with the severity of the hematological toxicities. Single-locus analysis showed that the single-nucleotide polymorphisms (SNPs) RRM1 rs12806698 and rs11031918 and DCTD rs7663494 were significantly associated with severe neutropenia, hENT1 rs760370 and hCNT3 rs7867504 and rs4877831 were associated with severe leukopenia, CDA rs2072671, DCTD rs7663494, and WEE1 rs3910384 were associated with severe anemia, and MTHFR rs1801133 was associated with severe thrombocytopenia after stringent Bonferroni correction (P < .0038). The gene-gene interaction analysis identified the overall best models, including a 2-way interaction model (hCNT3 rs7867504 and dCK rs12648166) for severe leukopenia (P = .0022) and a 3-locus model (CDA rs207671, DCTD rs7663494, and WEE1 rs3910384) for severe anemia with a strong synergistic effect (P = .0001). The association with hematological toxicity was further strengthened by the results of a haplotype analysis, in which the homozygous genotype combination of rs3910384 CC, rs2072671 AA, rs12648166 GG, rs7867504 CC, and rs7663494 TT conferred high genetic susceptibility to severe thrombocytopenia. Our results suggest that the gene-gene interaction of gemcitabine metabolic pathway genes and WEE1 contributes to susceptibility to gemcitabine-induced hematological toxicity. Moreover, we propose a promising data-mining analysis approach (generalized multifactor dimensionality reduction) to detect and characterize gene-gene interactions.
Collapse
Affiliation(s)
- Juan Li
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Bing Chen
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wen-Qi Xi
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Chen Yang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wei-Xia Zhang
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
6
|
Allard CAH, Opalko HE, Moseley JB. Stable Pom1 clusters form a glucose-modulated concentration gradient that regulates mitotic entry. eLife 2019; 8:e46003. [PMID: 31050341 PMCID: PMC6524964 DOI: 10.7554/elife.46003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/02/2019] [Indexed: 12/12/2022] Open
Abstract
Control of cell size requires molecular size sensors that are coupled to the cell cycle. Rod-shaped fission yeast cells divide at a threshold size partly due to Cdr2 kinase, which forms nodes at the medial cell cortex where it inhibits the Cdk1-inhibitor Wee1. Pom1 kinase phosphorylates and inhibits Cdr2, and forms cortical concentration gradients from cell poles. Pom1 inhibits Cdr2 signaling to Wee1 specifically in small cells, but the time and place of their regulatory interactions were unclear. We show that Pom1 forms stable oligomeric clusters that dynamically sample the cell cortex. Binding frequency is patterned into a concentration gradient by the polarity landmarks Tea1 and Tea4. Pom1 clusters colocalize with Cdr2 nodes, forming a glucose-modulated inhibitory threshold against node activation. Our work reveals how Pom1-Cdr2-Wee1 operates in multiprotein clusters at the cortex to promote mitotic entry at a cell size that can be modified by nutrient availability.
Collapse
Affiliation(s)
- Corey A H Allard
- Department of Biochemistry and Cell BiologyThe Geisel School of Medicine at DartmouthHanoverUnited States
| | - Hannah E Opalko
- Department of Biochemistry and Cell BiologyThe Geisel School of Medicine at DartmouthHanoverUnited States
| | - James B Moseley
- Department of Biochemistry and Cell BiologyThe Geisel School of Medicine at DartmouthHanoverUnited States
| |
Collapse
|
7
|
Allard CAH, Opalko HE, Liu KW, Medoh U, Moseley JB. Cell size-dependent regulation of Wee1 localization by Cdr2 cortical nodes. J Cell Biol 2018; 217:1589-1599. [PMID: 29514920 PMCID: PMC5940308 DOI: 10.1083/jcb.201709171] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 12/07/2017] [Accepted: 02/21/2018] [Indexed: 11/22/2022] Open
Abstract
Cell size control requires mechanisms that link cell growth with Cdk1 activity. In fission yeast, the protein kinase Cdr2 forms cortical nodes that include the Cdk1 inhibitor Wee1 along with the Wee1-inhibitory kinase Cdr1. We investigated how nodes inhibit Wee1 during cell growth. Biochemical fractionation revealed that Cdr2 nodes were megadalton structures enriched for activated Cdr2, which increases in level during interphase growth. In live-cell total internal reflection fluorescence microscopy videos, Cdr2 and Cdr1 remained constant at nodes over time, but Wee1 localized to nodes in short bursts. Recruitment of Wee1 to nodes required Cdr2 kinase activity and the noncatalytic N terminus of Wee1. Bursts of Wee1 localization to nodes increased 20-fold as cells doubled in size throughout G2. Size-dependent signaling was caused in part by the Cdr2 inhibitor Pom1, which suppressed Wee1 node bursts in small cells. Thus, increasing Cdr2 activity during cell growth promotes Wee1 localization to nodes, where inhibitory phosphorylation of Wee1 by Cdr1 and Cdr2 kinases promotes mitotic entry.
Collapse
Affiliation(s)
- Corey A H Allard
- Department of Biochemistry and Cell Biology, The Geisel School of Medicine at Dartmouth, Dartmouth, Hanover, NH
| | - Hannah E Opalko
- Department of Biochemistry and Cell Biology, The Geisel School of Medicine at Dartmouth, Dartmouth, Hanover, NH
| | - Ko-Wei Liu
- Department of Biochemistry and Cell Biology, The Geisel School of Medicine at Dartmouth, Dartmouth, Hanover, NH
| | - Uche Medoh
- Department of Biochemistry and Cell Biology, The Geisel School of Medicine at Dartmouth, Dartmouth, Hanover, NH
| | - James B Moseley
- Department of Biochemistry and Cell Biology, The Geisel School of Medicine at Dartmouth, Dartmouth, Hanover, NH
| |
Collapse
|
8
|
Abstract
Gerganova and Martin preview work from Allard et al. that describes the Wee1- and Cdr1/2-dependent mechanism by which cells link cell size with mitotic entry. All cells show size homeostasis owing to coordination of division with growth. In this issue, Allard et al. (2018. J. Cell Biol.https://doi.org/10.1083/jcb.201709171) establish that transient inhibitory visits of a negative regulator of Cdk1 to cortical oligomeric platforms increase in number and duration with cell growth, suggesting how Cdk1 activation is coupled to cell size.
Collapse
Affiliation(s)
- Veneta Gerganova
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Sophie G Martin
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
9
|
Lengefeld J, Hotz M, Rollins M, Baetz K, Barral Y. Budding yeast Wee1 distinguishes spindle pole bodies to guide their pattern of age-dependent segregation. Nat Cell Biol 2017; 19:941-951. [DOI: 10.1038/ncb3576] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 06/20/2017] [Indexed: 12/19/2022]
|
10
|
Lal S, Zarei M, Chand SN, Dylgjeri E, Mambelli-Lisboa NC, Pishvaian MJ, Yeo CJ, Winter JM, Brody JR. WEE1 inhibition in pancreatic cancer cells is dependent on DNA repair status in a context dependent manner. Sci Rep 2016; 6:33323. [PMID: 27616351 PMCID: PMC5018859 DOI: 10.1038/srep33323] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 08/23/2016] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a lethal disease, in part, because of the lack of effective targeted therapeutic options. MK-1775 (also known as AZD1775), a mitotic inhibitor, has been demonstrated to enhance the anti-tumor effects of DNA damaging agents such as gemcitabine. We evaluated the efficacy of MK-1775 alone or in combination with DNA damaging agents (MMC or oxaliplatin) in PDA cell lines that are either DNA repair proficient (DDR-P) or deficient (DDR-D). PDA cell lines PL11, Hs 766T and Capan-1 harboring naturally selected mutations in DNA repair genes FANCC, FANCG and BRCA2 respectively, were less sensitive to MK-1775 as compared to two out of four representative DDR-P (MIA PaCa2 and PANC-1) cell lines. Accordingly, DDR-P cells exhibit reduced sensitivity to MK-1775 upon siRNA silencing of DNA repair genes, BRCA2 or FANCD2, compared to control cells. Only DDR-P cells showed increased apoptosis as a result of early mitotic entry and catastrophe compared to DDR-D cells. Taken together with other recently published reports, our results add another level of evidence that the efficacy of WEE1 inhibition is influenced by the DNA repair status of a cell and may also be dependent on the tumor type and model evaluated.
Collapse
Affiliation(s)
- Shruti Lal
- Department of Surgery, Division of Surgical Research; Jefferson Pancreas, Biliary and Related Cancer Center; Jefferson Medical College; Thomas Jefferson University, Philadelphia, PA, USA
| | - Mahsa Zarei
- Department of Surgery, Division of Surgical Research; Jefferson Pancreas, Biliary and Related Cancer Center; Jefferson Medical College; Thomas Jefferson University, Philadelphia, PA, USA
| | - Saswati N Chand
- Department of Surgery, Division of Surgical Research; Jefferson Pancreas, Biliary and Related Cancer Center; Jefferson Medical College; Thomas Jefferson University, Philadelphia, PA, USA
| | - Emanuela Dylgjeri
- Department of Surgery, Division of Surgical Research; Jefferson Pancreas, Biliary and Related Cancer Center; Jefferson Medical College; Thomas Jefferson University, Philadelphia, PA, USA
| | - Nicole C Mambelli-Lisboa
- Department of Surgery, Division of Surgical Research; Jefferson Pancreas, Biliary and Related Cancer Center; Jefferson Medical College; Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael J Pishvaian
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 3800 Reservoir Road, NW, 20057 Washington DC, USA
| | - Charles J Yeo
- Department of Surgery, Division of Surgical Research; Jefferson Pancreas, Biliary and Related Cancer Center; Jefferson Medical College; Thomas Jefferson University, Philadelphia, PA, USA
| | - Jordan M Winter
- Department of Surgery, Division of Surgical Research; Jefferson Pancreas, Biliary and Related Cancer Center; Jefferson Medical College; Thomas Jefferson University, Philadelphia, PA, USA
| | - Jonathan R Brody
- Department of Surgery, Division of Surgical Research; Jefferson Pancreas, Biliary and Related Cancer Center; Jefferson Medical College; Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
11
|
Jang SH, Kim AR, Park NH, Park JW, Han IS. DRG2 Regulates G2/M Progression via the Cyclin B1-Cdk1 Complex. Mol Cells 2016; 39:699-704. [PMID: 27669826 PMCID: PMC5050535 DOI: 10.14348/molcells.2016.0149] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 02/01/2023] Open
Abstract
Developmentally regulated GTP-binding protein 2 (DRG2) plays an important role in cell growth. Here we explored the linkage between DRG2 and G2/M phase checkpoint function in cell cycle progression. We observed that knockdown of DRG2 in HeLa cells affected growth in a wound-healing assay, and tumorigenicity in nude mice xenografts. Flow cytometry assays and [(3)H] incorporation assays indicated that G2/M phase arrest was responsible for the decreased proliferation of these cells. Knockdown of DRG2 elicited down-regulation of the major mitotic promoting factor, the cyclin B1/Cdk1 complex, but up-regulation of the cell cycle arresting proteins, Wee1, Myt1, and p21. These findings identify a novel role of DRG2 in G2/M progression.
Collapse
Affiliation(s)
- Soo Hwa Jang
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Ah-Ram Kim
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - Neung-Hwa Park
- Department of Internal Medicine, University of Ulsan College of Medicine, Ulsan University Hospital, Ulsan 44033,
Korea
| | - Jeong Woo Park
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| | - In-Seob Han
- School of Biological Sciences, University of Ulsan, Ulsan 44610,
Korea
| |
Collapse
|
12
|
Jhuraney A, Woods NT, Wright G, Rix L, Kinose F, Kroeger JL, Remily-Wood E, Cress WD, Koomen JM, Brantley SG, Gray JE, Haura EB, Rix U, Monteiro AN. PAXIP1 Potentiates the Combination of WEE1 Inhibitor AZD1775 and Platinum Agents in Lung Cancer. Mol Cancer Ther 2016; 15:1669-81. [PMID: 27196765 PMCID: PMC4936941 DOI: 10.1158/1535-7163.mct-15-0182] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 04/09/2016] [Indexed: 11/16/2022]
Abstract
The DNA damage response (DDR) involves a complex network of signaling events mediated by modular protein domains such as the BRCA1 C-terminal (BRCT) domain. Thus, proteins that interact with BRCT domains and are a part of the DDR constitute potential targets for sensitization to DNA-damaging chemotherapy agents. We performed a pharmacologic screen to evaluate 17 kinases, identified in a BRCT-mediated interaction network as targets to enhance platinum-based chemotherapy in lung cancer. Inhibition of mitotic kinase WEE1 was found to have the most effective response in combination with platinum compounds in lung cancer cell lines. In the BRCT-mediated interaction network, WEE1 was found in complex with PAXIP1, a protein containing six BRCT domains involved in transcription and in the cellular response to DNA damage. We show that PAXIP1 BRCT domains regulate WEE1-mediated phosphorylation of CDK1. Furthermore, ectopic expression of PAXIP1 promotes enhanced caspase-3-mediated apoptosis in cells treated with WEE1 inhibitor AZD1775 (formerly, MK-1775) and cisplatin compared with cells treated with AZD1775 alone. Cell lines and patient-derived xenograft models expressing both PAXIP1 and WEE1 exhibited synergistic effects of AZD1775 and cisplatin. In summary, PAXIP1 is involved in sensitizing lung cancer cells to the WEE1 inhibitor AZD1775 in combination with platinum-based treatment. We propose that WEE1 and PAXIP1 levels may be used as mechanism-based biomarkers of response when WEE1 inhibitor AZD1775 is combined with DNA-damaging agents. Mol Cancer Ther; 15(7); 1669-81. ©2016 AACR.
Collapse
Affiliation(s)
- Ankita Jhuraney
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida. Cancer Biology PhD Program, University of South Florida, Tampa, Florida
| | - Nicholas T Woods
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Gabriela Wright
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Lily Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Fumi Kinose
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jodi L Kroeger
- Flow Cytometry Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Elizabeth Remily-Wood
- Molecular Oncology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - W Douglas Cress
- Molecular Oncology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - John M Koomen
- Molecular Oncology Program, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Stephen G Brantley
- M2Gen, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida.
| | - Alvaro N Monteiro
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, Florida.
| |
Collapse
|
13
|
Liu D, Wu C, Jiao Y, Hou L, Lu D, Zheng H, Chen C, Qian J, Fei K, Su B. WEE1 kinase polymorphism as a predictive biomarker for efficacy of platinum-gemcitabine doublet chemotherapy in advanced non-small cell lung cancer patients. Sci Rep 2015; 5:11114. [PMID: 26057002 PMCID: PMC4460872 DOI: 10.1038/srep11114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 05/11/2015] [Indexed: 12/31/2022] Open
Abstract
DNA-damaging agents are commonly used for first-line chemotherapy of advanced non-small cell lung cancer (NSCLC). As a G2/M checkpoint kinase, Wee1 can phosphorylate CDC2-tyr15 and induce G2/M cell cycle arrest in response to DNA damage. The correlation of WEE1 polymorphisms to the efficacy of chemotherapy was tested in 663 advanced NSCLC patients. WEE1 rs3910384 genotype correlated to overall survival (OS) and progress-free survival (PFS) of NSCLC patients treated with platinum-based chemotherapy. Sub-group analysis revealed that rs3910384 was particularly associated with the efficacy of doublet chemotherapy combining two DNA-damaging agents, i.e. platinum and gemcitabine. NSCLC patients with the WEE1 rs3910384 G/G homozygote genotype showed 13.5 months extended OS, 3.2 months extended PFS, and a 274% relative increase in their 3-year survival rate (from 7.4% to 27.7%) compared to the A/A+A/G genotype after treatment with platinum-gemcitabine regimen. This finding was reproduced in the validation cohort. We utilized a luciferase reporter assay and Electrophoretic Mobility Shift Assay (EMSA) to demonstrate that rs3910384-linked WEE1 promoter haplotype can mediate allele-specific transcriptional binding and WEE1 expression in DNA damage response. In conclusion, the WEE1 rs3910384 G/G homozygote genotype can be used as a selective biomarker for NSCLC patients to indicate treatment with platinum and gemcitabine regimen.
Collapse
Affiliation(s)
| | | | | | | | - Daru Lu
- Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Hui Zheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine
| | - Ji Qian
- Fudan University Shanghai Cancer Center, State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Ke Fei
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine
| | | |
Collapse
|
14
|
Wang YX, Cai H, Jiang G, Zhou TB, Wu H. Silibinin Inhibits Proliferation, Induces Apoptosis and Causes Cell Cycle Arrest in Human Gastric Cancer MGC803 Cells Via STAT3 Pathway Inhibition. Asian Pac J Cancer Prev 2014; 15:6791-8. [DOI: 10.7314/apjcp.2014.15.16.6791] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
15
|
Effects of human fibronectin and human serum albumin sequential adsorption on preosteoblastic cell adhesion. Biointerphases 2014; 9:029008. [DOI: 10.1116/1.4867598] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
16
|
Lal S, Burkhart RA, Beeharry N, Bhattacharjee V, Londin ER, Cozzitorto JA, Romeo C, Jimbo M, Norris ZA, Yeo CJ, Sawicki JA, Winter JM, Rigoutsos I, Yen TJ, Brody JR. HuR posttranscriptionally regulates WEE1: implications for the DNA damage response in pancreatic cancer cells. Cancer Res 2014; 74:1128-40. [PMID: 24536047 DOI: 10.1158/0008-5472.can-13-1915] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
HuR (ELAV1), an RNA-binding protein abundant in cancer cells, primarily resides in the nucleus, but under specific stress (e.g., gemcitabine), HuR translocates to the cytoplasm in which it tightly modulates the expression of mRNA survival cargo. Here, we demonstrate for the first time that stressing pancreatic ductal adenocarcinoma (PDA) cells by treatment with DNA-damaging anticancer agents (mitomycin C, oxaliplatin, cisplatin, carboplatin, and a PARP inhibitor) results in HuR's translocation from the nucleus to the cytoplasm. Importantly, silencing HuR in PDA cells sensitized the cells to these agents, whereas overexpressing HuR caused resistance. HuR's role in the efficacy of DNA-damaging agents in PDA cells was, in part, attributed to the acute upregulation of WEE1 by HuR. WEE1, a mitotic inhibitor kinase, regulates the DNA damage repair pathway, and therapeutic inhibition of WEE1 in combination with chemotherapy is currently in early phase trials for the treatment of cancer. We validate WEE1 as a HuR target in vitro and in vivo by demonstrating (i) direct binding of HuR to WEE1's mRNA (a discrete 56-bp region residing in the 3' untranslated region) and (ii) HuR siRNA silencing and overexpression directly affects the protein levels of WEE1, especially after DNA damage. HuR's positive regulation of WEE1 increases γ-H2AX levels, induces Cdk1 phosphorylation, and promotes cell-cycle arrest at the G2-M transition. We describe a novel mechanism that PDA cells use to protect against DNA damage in which HuR posttranscriptionally regulates the expression and downstream function of WEE1 upon exposure to DNA-damaging agents.
Collapse
Affiliation(s)
- Shruti Lal
- Authors' Affiliations: Department of Surgery, Division of Surgical Research, Jefferson Pancreas, Biliary and Related Cancer Center, Jefferson Medical College; Computational Medicine Center; Kimmel Cancer Center, Thomas Jefferson University; Fox Chase Cancer Center, Philadelphia; and Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Pan KZ, Saunders TE, Flor-Parra I, Howard M, Chang F. Cortical regulation of cell size by a sizer cdr2p. eLife 2014; 3:e02040. [PMID: 24642412 PMCID: PMC3956294 DOI: 10.7554/elife.02040] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/12/2014] [Indexed: 01/03/2023] Open
Abstract
Cells can, in principle, control their size by growing to a specified size before commencing cell division. How any cell actually senses its own size remains poorly understood. The fission yeast Schizosaccharomyces pombe are rod-shaped cells that grow to ∼14 µm in length before entering mitosis. In this study, we provide evidence that these cells sense their surface area as part of this size control mechanism. We show that cells enter mitosis at a certain surface area, as opposed to a certain volume or length. A peripheral membrane protein kinase cdr2p has properties of a dose-dependent 'sizer' that controls mitotic entry. As cells grow, the local cdr2p concentration in nodes at the medial cortex accumulates as a measure of cell surface area. Our findings, which challenge a previously proposed pom1p gradient model, lead to a new model in which cells sense their size by using cdr2p to probe the surface area over the whole cell and relay this information to the medial cortex. DOI: http://dx.doi.org/10.7554/eLife.02040.001.
Collapse
Affiliation(s)
- Kally Z Pan
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, United States
| | - Timothy E Saunders
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratories, Heidelberg, Germany
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Ignacio Flor-Parra
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, United States
| | - Martin Howard
- Computational and Systems Biology, John Innes Centre, Norwich, United Kingdom
| | - Fred Chang
- Department of Microbiology and Immunology, Columbia University College of Physicians and Surgeons, New York, United States
| |
Collapse
|
18
|
Abstract
The activation of the Cdk1 (cyclin-dependent kinase 1)-cyclin B complex to promote commitment to mitosis is controlled by the phosphorylation status of the Cdk1 catalytic subunit. Cdk1 phosphorylation by Wee1 kinases blocks activation until Cdc25 (cell division cycle 25) phosphatases remove this phosphate to drive division. Feedback inhibition of Wee1 and promotion of Cdc25 activities by the newly activated Cdk1-cyclin B complexes ensure that the transition from interphase to mitosis is a rapid and complete bi-stable switch. Although this level of molecular understanding of the mitotic commitment switch has been clear for over two decades, it is still unclear how the switch is engaged to promote division at the right time for a particular context. We discuss recent work in fission yeast that shows how the spatial organization of signalling networks, in particular events on the centrosome equivalent, the spindle pole body, plays a key role in ensuring that the timing of cell division is coupled to environmental cues.
Collapse
Affiliation(s)
- Iain M Hagan
- *Cell Division Group, CRUK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, U.K
| | | |
Collapse
|
19
|
Epigenetically induced paucity of histone H2A.Z stabilizes fission-yeast ectopic centromeres. Nat Struct Mol Biol 2013; 20:1397-406. [PMID: 24186062 DOI: 10.1038/nsmb.2697] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 09/16/2013] [Indexed: 11/08/2022]
Abstract
In most eukaryotes, centromeres are epigenetically defined by nucleosomes that contain the histone H3 variant centromere protein A (CENP-A). Specific targeting of the CENP-A-loading chaperone to the centromere is vital for stable centromere propagation; however, the existence of ectopic centromeres (neocentromeres) indicates that this chaperone can function in different chromatin environments. The mechanism responsible for accommodating the CENP-A chaperone at noncentromeric regions is poorly understood. Here, we report the identification of transient, immature neocentromeres in Schizosaccharomyces pombe that show reduced association with the CENP-A chaperone Scm3, owing to persistence of the histone H2A variant H2A.Z. After the acquisition of adjacent heterochromatin or relocation of the immature neocentromeres to subtelomeric regions, H2A.Z was depleted and Scm3 was replenished, thus leading to subsequent stabilization of the neocentromeres. These findings provide new insights into histone variant-mediated epigenetic control of neocentromere establishment.
Collapse
|
20
|
Tay YD, Patel A, Kaemena DF, Hagan IM. Mutation of a conserved residue enhances the sensitivity of analogue-sensitised kinases to generate a novel approach to the study of mitosis in fission yeast. J Cell Sci 2013; 126:5052-61. [PMID: 23986474 DOI: 10.1242/jcs.135301] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The chemical genetic strategy in which mutational enlargement of the ATP-binding site sensitises of a protein kinase to bulky ATP analogues has proved to be an elegant tool for the generation of conditional analogue-sensitive kinase alleles in a variety of model organisms. Here, we describe a novel substitution mutation in the kinase domain that can enhance the sensitivity of analogue-sensitive kinases. Substitution of a methionine residue to phenylalanine in the +2 position after HRDLKxxN motif of the subdomain VIb within the kinase domain markedly increased the sensitivities of the analogue-sensitive kinases to ATP analogues in three out of five S. pombe kinases (i.e. Plo1, Orb5 and Wee1) that harbor this conserved methionine residue. Kinome alignment established that a methionine residue is found at this site in 5-9% of kinases in key model organisms, suggesting that a broader application of this structural modification may enhance ATP analogue sensitivity of analogue-sensitive kinases in future studies. We also show that the enhanced sensitivity of the wee1.as8 allele in a cdc25.22 background can be exploited to generate highly synchronised mitotic and S phase progression at 36°C. Proof-of-principle experiments show how this novel synchronisation technique will prove of great use in the interrogation of the mitotic or S-phase functions through temperature sensitivity mutation of molecules of interest in fission yeast.
Collapse
Affiliation(s)
- Ye-Dee Tay
- CRUK Cell Division Group, Paterson Institute for Cancer Research, Wilmslow Road, Manchester M20 4BX, UK
| | | | | | | |
Collapse
|
21
|
Roy RV, Suman S, Das TP, Luevano J, Damodaran C. Withaferin A, a steroidal lactone from Withania somnifera, induces mitotic catastrophe and growth arrest in prostate cancer cells. JOURNAL OF NATURAL PRODUCTS 2013; 76:1909-15. [PMID: 24079846 PMCID: PMC4144448 DOI: 10.1021/np400441f] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Cell cycle deregulation is strongly associated with the pathogenesis of prostate cancer. Clinical trials of cell cycle regulators that target either the G0/G1 or G2/M phase to inhibit the growth of cancers including prostate cancer are increasing. The present study focused on the cell cycle regulatory potential of the withanolide withaferin A (1) on prostate cancer cells. Compound 1 induced G2/M arrest in both prostate cancer cell lines (PC-3 and DU-145) when treated for 48 h. The G2/M arrest was accompanied by upregulation of phosphorylated Wee-1, phosphorylated histone H3, p21, and Aurora B. On the other hand, downregulation of cyclins (A2, B1, and E2) and a reduction in phosphorylated Cdc2 (Tyr15) were observed in 1-treated prostate cancer cells. In addition, decreased levels of phosphorylated Chk1 (Ser345) and Chk2 (Thr68) were evident in prostate cancer cells on treatment with 1. These results suggest that activation of Cdc2 leads to arrest in the M phase, with abnormal duplication, and initiation of mitotic catastrophe that results in cell death. In conclusion, these results show clearly the potential of 1 as a regulator of the G2/M phase of the cell cycle and as a therapeutic agent for prostate cancer.
Collapse
Affiliation(s)
| | | | | | | | - Chendil Damodaran
- Chendil Damodaran, Center of Excellence in Cancer Research, Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX 79905, , Phone: 915-215-4228, Fax: 915-783-522
| |
Collapse
|
22
|
Ji T, Lin C, Krill LS, Eskander R, Guo Y, Zi X, Hoang BH. Flavokawain B, a kava chalcone, inhibits growth of human osteosarcoma cells through G2/M cell cycle arrest and apoptosis. Mol Cancer 2013; 12:55. [PMID: 23764122 PMCID: PMC3681603 DOI: 10.1186/1476-4598-12-55] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 05/30/2013] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary bone malignancy with a high propensity for local invasion and distant metastasis. Limited by the severe toxicity of conventional agents, the therapeutic bottleneck of osteosarcoma still remains unconquered. Flavokawain B (FKB), a kava extract, has been reported to have significant anti-tumor effects on several carcinoma cell lines both in vitro and in vivo. Its efficacy and low toxicity profile make FKB a promising agent for use as a novel chemotherapeutic agent. RESULTS In the current study, we investigated the anti-proliferative and apoptotic effects of FKB against human osteosarcomas. Exposure of OS cells to FKB resulted in apoptosis, evidenced by loss of cell viability, morphological changes and the externalization of phosphatidylserine. Apoptosis induced by FKB resulted in activation of Caspase-3/7, -8 and -9 in OS cell lines, 143B and Saos-2. FKB also down-regulated inhibitory apoptotic markers, including Bcl-2 and Survivin and led to concomitant increases in apoptotic proteins, Bax, Puma and Fas. Therefore, the induction of apoptosis by FKB involved both extrinsic and intrinsic pathways. FKB also caused G2/M phase cell cycle arrest, which was observed through reductions in the levels of cyclin B1, cdc2 and cdc25c and increases in Myt1 levels. Furthermore, migration and invasion ability was decreased by FKB in a dose-dependent manner. The cytotoxicity profile showed FKB had significant lower side effects on bone marrow cells and small intestinal epithelial cells compared with Adriamycin. CONCLUSIONS Taken together, our evidence of apoptosis and cell cycle arrest by FKB treatment with less toxicity than the standard treatments provides an innovative argument for the use of FKB as a chemotherapeutic and chemopreventive compound. In vivo experiments utilizing FKB to reduce tumorigenesis and metastatic potential will be crucial to further justify clinical application.
Collapse
Affiliation(s)
- Tao Ji
- Department of Orthopaedic Surgery, UC Irvine Multidisciplinary Sarcoma Center, Chao Family Comprehensive Cancer Center, University of California, Irvine, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Fleißner A. Turning the switch: using chemical genetics to elucidate protein kinase functions in filamentous fungi. FUNGAL BIOL REV 2013. [DOI: 10.1016/j.fbr.2013.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
24
|
Effects of light illumination and the expression of wee1 on tissue regeneration in adult zebrafish. Biochem Biophys Res Commun 2012; 428:132-6. [DOI: 10.1016/j.bbrc.2012.10.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 10/03/2012] [Indexed: 12/21/2022]
|
25
|
Lee IJ, Coffman VC, Wu JQ. Contractile-ring assembly in fission yeast cytokinesis: Recent advances and new perspectives. Cytoskeleton (Hoboken) 2012; 69:751-63. [PMID: 22887981 DOI: 10.1002/cm.21052] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 07/16/2012] [Indexed: 11/07/2022]
Abstract
The fission yeast Schizosaccharomyces pombe is an excellent model organism to study cytokinesis. Here, we review recent advances on contractile-ring assembly in fission yeast. First, we summarize the assembly of cytokinesis nodes, the precursors of a normal contractile ring. IQGAP Rng2 and myosin essential light chain Cdc4 are recruited by the anillin-like protein Mid1, followed by the addition of other cytokinesis node proteins. Mid1 localization on the plasma membrane is stabilized by interphase node proteins. Second, we discuss proteins and processes that contribute to the search, capture, pull, and release mechanism of contractile-ring assembly. Actin filaments nucleated by formin Cdc12, the motor activity of myosin-II, the stiffness of the actin network, and severing of actin filaments by cofilin all play essential roles in contractile-ring assembly. Finally, we discuss the Mid1-independent pathway for ring assembly, and the possible mechanisms underlying the ring maturation and constriction. Collectively, we provide an overview of the current understanding of contractile-ring assembly and uncover future directions in studying cytokinesis in fission yeast.
Collapse
Affiliation(s)
- I-Ju Lee
- Department of Molecular Genetics, The Ohio State University, Columbus, Ohio, USA
| | | | | |
Collapse
|
26
|
Hiraoka Y, Maekawa H, Asakawa H, Chikashige Y, Kojidani T, Osakada H, Matsuda A, Haraguchi T. Inner nuclear membrane protein Ima1 is dispensable for intranuclear positioning of centromeres. Genes Cells 2011; 16:1000-11. [DOI: 10.1111/j.1365-2443.2011.01544.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
27
|
Lv H, Yang YX, Zhang LD, Bai YQ. Relationship between WEE 1 and hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2011; 19:1515-1519. [DOI: 10.11569/wcjd.v19.i14.1515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of WEE 1 in hepatocellular carcinoma (HCC) and to analyze its relationship with clinicopathological characteristics of HCC.
METHODS: Twenty-three normal human liver tissue specimens, 20 cirrhosis specimens, and 42 HCC specimens were used in this study. Reverse transcriptional-polymerase chain reaction (RT-PCR) was used to measure the expression of WEE 1 mRNA in the above tissue specimens, while Western blot and immunohistochemistry were used to detect the expression of WEE 1 protein. The relationship between WEE 1 expression and clinicopathological characteristics of HCC was analyzed.
RESULTS: The positive rates of Wee 1 mRNA expression in normal liver tissue, cirrhosis and HCC were 21.7%, 55% and 90.5%, respectively, with a significant difference among the three groups (P < 0.01). The positive rates of Wee 1 protein expression as revealed by Western blot and immunohistochemistry in the above three groups were 13.04%/17.4%, 40%/60% and 78.6%/83.3%, respectively, with significant differences among the three groups (both P < 0.01). Up-regulated expression of WEE 1 was significantly correlated with tumor differentiation and pathological grade in HCC (χ2 = 17.454, P < 0.01; χ2 = 14.559, P < 0.01).
CONCLUSION: WEE 1 expression was significantly up-regulated in HCC. High expression of WEE 1 may be closely related with tumor differentiation and pathological grade in HCC.
Collapse
|