1
|
El Dika M, Dudka D, Kloc M, Kubiak JZ. CDC6 as a Key Inhibitory Regulator of CDK1 Activation Dynamics and the Timing of Mitotic Entry and Progression. BIOLOGY 2023; 12:855. [PMID: 37372141 DOI: 10.3390/biology12060855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023]
Abstract
Timely mitosis is critically important for early embryo development. It is regulated by the activity of the conserved protein kinase CDK1. The dynamics of CDK1 activation must be precisely controlled to assure physiologic and timely entry into mitosis. Recently, a known S-phase regulator CDC6 emerged as a key player in mitotic CDK1 activation cascade in early embryonic divisions, operating together with Xic1 as a CDK1 inhibitor upstream of the Aurora A and PLK1, both CDK1 activators. Herein, we review the molecular mechanisms that underlie the control of mitotic timing, with special emphasis on how CDC6/Xic1 function impacts CDK1 regulatory network in the Xenopus system. We focus on the presence of two independent mechanisms inhibiting the dynamics of CDK1 activation, namely Wee1/Myt1- and CDC6/Xic1-dependent, and how they cooperate with CDK1-activating mechanisms. As a result, we propose a comprehensive model integrating CDC6/Xic1-dependent inhibition into the CDK1-activation cascade. The physiological dynamics of CDK1 activation appear to be controlled by the system of multiple inhibitors and activators, and their integrated modulation ensures concomitantly both the robustness and certain flexibility of the control of this process. Identification of multiple activators and inhibitors of CDK1 upon M-phase entry allows for a better understanding of why cells divide at a specific time and how the pathways involved in the timely regulation of cell division are all integrated to precisely tune the control of mitotic events.
Collapse
Affiliation(s)
- Mohammed El Dika
- Department of Biochemistry, Larner College of Medicine, UVM Cancer Center, University of Vermont, Burlington, VT 05405, USA
| | - Damian Dudka
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Transplant Immunology, Houston, TX 77030, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX 77030, USA
- Department of Genetics, MD Anderson Cancer Center, The University of Texas, Houston, TX 77030, USA
| | - Jacek Z Kubiak
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine-National Research Institute (WIM-PIB), Szaserow 128, 04-141 Warsaw, Poland
- Dynamics and Mechanics of Epithelia Group, Faculty of Medicine, Institute of Genetics and Development of Rennes, University of Rennes, CNRS, UMR 6290, 35043 Rennes, France
| |
Collapse
|
2
|
Siebert A, Gattringer V, Weishaupt JH, Behrends C. ALS-linked loss of Cyclin-F function affects HSP90. Life Sci Alliance 2022; 5:5/12/e202101359. [PMID: 36114006 PMCID: PMC9481933 DOI: 10.26508/lsa.202101359] [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: 12/30/2021] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Analysis of ALS patient cell lines and cyclin-F overexpression and knockout cells identified HSP90AB1 as novel SCFcyclin-F substrate pointing to a loss-of-function mechanism for ALS CCNF mutations. The founding member of the F-box protein family, Cyclin-F, serves as a substrate adaptor for the E3 ligase Skp1-Cul1-F-box (SCF)Cyclin-F which is responsible for ubiquitination of proteins involved in cell cycle progression, DNA damage and mitotic fidelity. Missense mutations in CCNF encoding for Cyclin-F are associated with amyotrophic lateral sclerosis (ALS). However, it remains elusive whether CCNF mutations affect the substrate adaptor function of Cyclin-F and whether altered SCFCyclin-F–mediated ubiquitination contributes to pathogenesis in CCNF mutation carriers. To address these questions, we set out to identify new SCFCyclin-F targets in neuronal and ALS patient–derived cells. Mass spectrometry–based ubiquitinome profiling of CCNF knockout and mutant cell lines as well as Cyclin-F proximity and interaction proteomics converged on the HSP90 chaperone machinery as new substrate candidate. Biochemical analyses provided evidence for a Cyclin-F–dependent association and ubiquitination of HSP90AB1 and implied a regulatory role that could affect the binding of a number of HSP90 clients and co-factors. Together, our results point to a possible Cyclin-F loss-of-function–mediated chaperone dysregulation that might be relevant for ALS.
Collapse
Affiliation(s)
- Alexander Siebert
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Vanessa Gattringer
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Jochen H Weishaupt
- Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, Mannheim, Germany
| | - Christian Behrends
- Munich Cluster for Systems Neurology (SyNergy), Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| |
Collapse
|
3
|
Understanding the Underlying Molecular Mechanisms of Meiotic Arrest during In Vitro Spermatogenesis in Rat Prepubertal Testicular Tissue. Int J Mol Sci 2022; 23:ijms23115893. [PMID: 35682573 PMCID: PMC9180380 DOI: 10.3390/ijms23115893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/18/2022] [Accepted: 05/22/2022] [Indexed: 12/10/2022] Open
Abstract
In vitro spermatogenesis appears to be a promising approach to restore the fertility of childhood cancer survivors. The rat model has proven to be challenging, since germ cell maturation is arrested in organotypic cultures. Here, we report that, despite a meiotic entry, abnormal synaptonemal complexes were found in spermatocytes, and in vitro matured rat prepubertal testicular tissues displayed an immature phenotype. RNA-sequencing analyses highlighted up to 600 differentially expressed genes between in vitro and in vivo conditions, including genes involved in blood-testis barrier (BTB) formation and steroidogenesis. BTB integrity, the expression of two steroidogenic enzymes, and androgen receptors were indeed altered in vitro. Moreover, most of the top 10 predicted upstream regulators of deregulated genes were involved in inflammatory processes or immune cell recruitment. However, none of the three anti-inflammatory molecules tested in this study promoted meiotic progression. By analysing for the first time in vitro matured rat prepubertal testicular tissues at the molecular level, we uncovered the deregulation of several genes and revealed that defective BTB function, altered steroidogenic pathway, and probably inflammation, could be at the origin of meiotic arrest.
Collapse
|
4
|
Cristea I, Bruland O, Aukrust I, Rødahl E, Bredrup C. Pellino-2 in nonimmune cells: novel interaction partners and intracellular localization. FEBS Lett 2021; 595:2909-2921. [PMID: 34674267 DOI: 10.1002/1873-3468.14212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/04/2021] [Accepted: 10/14/2021] [Indexed: 01/18/2023]
Abstract
Pellino-2 is an E3 ubiquitin ligase that mediates intracellular signaling in innate immune pathways. Most studies of endogenous Pellino-2 have been performed in macrophages, but none in nonimmune cells. Using yeast two-hybrid screening and co-immunoprecipitation, we identified six novel interaction partners of Pellino-2, with various localizations: insulin receptor substrate 1, NIMA-related kinase 9, tumor necrosis factor receptor-associated factor 7, cyclin-F, roundabout homolog 1, and disheveled homolog 2. Pellino-2 showed cytoplasmic localization in a wide range of nonimmune cells under physiological potassium concentrations. Treatment with the potassium ionophore nigericin resulted in nuclear localization of Pellino-2, which was reversed by the potassium channel blocker tetraethylammonium. Live-cell imaging revealed intracellular migration of GFP-tagged Pellino-2. In summary, Pellino-2 interacts with proteins at different cellular locations, taking part in dynamic processes that change its intracellular localization influenced by potassium efflux.
Collapse
Affiliation(s)
- Ileana Cristea
- Department of Clinical Medicine, University of Bergen, Norway
| | - Ove Bruland
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Ingvild Aukrust
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Norway
| | - Eyvind Rødahl
- Department of Clinical Medicine, University of Bergen, Norway.,Department of Ophthalmology, Haukeland University Hospital, Bergen, Norway
| | - Cecilie Bredrup
- Department of Clinical Medicine, University of Bergen, Norway.,Department of Ophthalmology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
5
|
Chang SC, Hung CS, Zhang BX, Hsieh TH, Hsu W, Ding JL. A Novel Signature of CCNF-Associated E3 Ligases Collaborate and Counter Each Other in Breast Cancer. Cancers (Basel) 2021; 13:cancers13122873. [PMID: 34201347 PMCID: PMC8228695 DOI: 10.3390/cancers13122873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/03/2021] [Accepted: 06/05/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary The dysregulation of UPS exacerbates the tumor microenvironment and drives malignant transformation. As the largest family of E3 ligases, the SCFF-boxes promotes BRCA progression. FBXL8 was recently identified to be a novel SCF E3 ligase that potently promotes BRCA. Here, we profiled the transcriptome of BRCA patient tissues by global NGS RNA-Seq and TCGA database analyses. A signature of four SCFF-box E3 ligases (FBXL8, FBXO43, FBXO15, CCNF) was found to be pivotal for BRCA advancement. Knockdown of FBXL8 and FBXO43 reduced cancer cell viability and proliferation, suggesting their pro-tumorigenic roles. However, the overexpression of CCNF inhibited cancer cell progression, indicating its anti-tumorigenic role. FBXL8 and FZR1 pulled down CCNF, and double knockdown of FBXL8 and FZR1 caused CCNF accumulation. Additionally, CCNF partnered with a pro-tumorigenic factor, RRM2, and overexpression of CCNF reduced RRM2. Our findings suggest a potential for drugging CCNF in co-modulatory partnership with FBXL8 and FZR1, for anti-BRCA therapy. Abstract Breast cancer (BRCA) malignancy causes major fatalities amongst women worldwide. SCF (Skp1-cullin-F-box proteins) E3 ubiquitin ligases are the most well-known members of the ubiquitination–proteasome system (UPS), which promotes cancer initiation and progression. Recently, we demonstrated that FBXL8, a novel F-box protein (SCFF-boxes) of SCF E3 ligase, accelerates BRCA advancement and metastasis. Since SCFF-boxes is a key component of E3 ligases, we hypothesized that other SCFF-boxes besides FBXL8 probably collaborate in regulating breast carcinogenesis. In this study, we retrospectively profiled the transcriptome of BRCA tissues and found a notable upregulation of four SCFF-box E3 ligases (FBXL8, FBXO43, FBXO15, and CCNF) in the carcinoma tissues. Similar to FBXL8, the knockdown of FBXO43 reduced cancer cell viability and proliferation, suggesting its pro-tumorigenic role. The overexpression of CCNF inhibited cancer cell progression, indicating its anti-tumorigenic role. Unexpectedly, CCNF protein was markedly downregulated in BRCA tissues, although its mRNA level was high. We showed that both E3 ligases, FBXL8 and FZR1, pulled down CCNF. Double knockdown of FBXL8 and FZR1 caused CCNF accumulation. On the other hand, CCNF itself pulled down a tumorigenic factor, RRM2, and CCNF overexpression reduced RRM2. Altogether, we propose a signature network of E3 ligases that collaboratively modulates CCNF anti-cancer activity. There is potential to target BRCA through modulation of the partnership axes of (i) CCNF-FBXL8, (ii) CCNF-FZR1, and (iii) CCNF-RRM2, particularly, via CCNF overexpression and activation and FBXL8/FZR1 suppression.
Collapse
Affiliation(s)
- Shu-Chun Chang
- The Ph.D. Program for Translational Medicine, College for Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- Correspondence: (S.-C.C.); (W.H.); (J.L.D.)
| | - Chin-Sheng Hung
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Division of General Surgery, Department of Surgery, Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare, New Taipei City 23561, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Bo-Xiang Zhang
- The Ph.D. Program for Translational Medicine, College for Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan;
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Tsung-Han Hsieh
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 110, Taiwan;
| | - Wayne Hsu
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Correspondence: (S.-C.C.); (W.H.); (J.L.D.)
| | - Jeak Ling Ding
- Department of Biological Sciences, National University of Singapore, Singapore 117543, Singapore
- Correspondence: (S.-C.C.); (W.H.); (J.L.D.)
| |
Collapse
|
6
|
Gray S, Santiago ER, Chappie JS, Cohen PE. Cyclin N-Terminal Domain-Containing-1 Coordinates Meiotic Crossover Formation with Cell-Cycle Progression in a Cyclin-Independent Manner. Cell Rep 2021; 32:107858. [PMID: 32640224 PMCID: PMC7341696 DOI: 10.1016/j.celrep.2020.107858] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/14/2020] [Accepted: 06/12/2020] [Indexed: 01/01/2023] Open
Abstract
During mammalian meiotic prophase I, programmed DNA double-strand breaks are repaired by non-crossover or crossover events, the latter predominantly occurring via the class I crossover pathway and requiring the cyclin N-terminal domain-containing 1(CNTD1) protein. Using an epitope-tagged Cntd1 allele, we detect a short isoform of CNTD1 in vivo that lacks a predicted N-terminal cyclin domain and does not bind cyclin-dependent kinases. Instead, we find that the short-form CNTD1 variant associates with components of the replication factor C (RFC) machinery to facilitate crossover formation, and with the E2 ubiquitin conjugating enzyme, CDC34, to regulate ubiquitylation and subsequent degradation of the WEE1 kinase, thereby modulating cell-cycle progression. We propose that these interactions facilitate a role for CNTD1 as a stop-go regulator during prophase I, ensuring accurate and complete crossover formation before allowing metaphase progression and the first meiotic division. CNTD1 associates with sites of crossing over in meiosis, co-localizing with MutLγ In the testis, CNTD1 does not interact with CDKs or with known crossover regulators CNTD1 regulates crossing over via interactions with the replication factor C complex CNTD1 regulates cell-cycle progression via interactions with the SCF complex
Collapse
Affiliation(s)
- Stephen Gray
- Department of Biomedical Sciences and Center for Reproductive Genomics, Cornell University, Ithaca, NY 14853, USA.
| | - Emerson R Santiago
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Joshua S Chappie
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Paula E Cohen
- Department of Biomedical Sciences and Center for Reproductive Genomics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
7
|
Yang M, Hu C, Cao Y, Liang W, Yang X, Xiao T. Ursolic Acid Regulates Cell Cycle and Proliferation in Colon Adenocarcinoma by Suppressing Cyclin B1. Front Pharmacol 2021; 11:622212. [PMID: 33628185 PMCID: PMC7898669 DOI: 10.3389/fphar.2020.622212] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/04/2020] [Indexed: 01/20/2023] Open
Abstract
Aims: The biological functions of cyclin B1 (CCNB1) in colon adenocarcinoma (COAD) will be explored in this study. Furthermore, the therapeutic effects and potential molecular mechanisms of ursolic acid (UA) in COAD cells will also be investigated in vitro. Methods: COAD data were obtained from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. Differentially expressed genes (DEGs) were determined with differential analysis. The biological functions of CCNB1 were analyzed through the GeneCards, the Search Tool for the Retrieval of Interacting Genes (STRING), and the Database for Annotation, Visualization, and Integrated Discovery (DAVID) databases. Therapeutic effects of UA on COAD cell lines HCT-116 and SW-480 were analyzed by CCK-8 and high-content screening (HCS) imaging assay. Flow cytometry was utilized to detect cell cycle changes of SW-480 and HCT-116 cells. Levels of mRNA and expression proteins of HCT-116, SW-480, and normal colon epithelial cells NCM-460 were determined by qRT-PCR and western blot. Results: CCNB1 was highly expressed and acted as an oncogene in COAD patients. CCNB1 and its interacting genes were significantly enriched in the cell cycle pathway. UA effectively inhibited the proliferation and injured COAD cells. In addition, UA arrested cell cycle of COAD cells in S phase. With regard to the molecular mechanisms of UA, we demonstrated that UA can significantly downregulate CCNB1 and its interacting genes and proteins, including CDK1, CDC20, CCND1, and CCNA2, which contributed to cell cycle blocking and COAD treatment. Conclusion: Results from this study revealed that UA possesses therapeutic effects on COAD. The anti-COAD activities of UA are tightly related to suppression of CCNB1 and its interacting targets, which is crucial in abnormal cell cycle process.
Collapse
Affiliation(s)
- Minhui Yang
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Changxiao Hu
- College of Clinical Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yibo Cao
- Colorectal and Anal Surgery, the First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu, China
| | - Wanling Liang
- Colorectal and Anal Surgery, the First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiangdong Yang
- Colorectal and Anal Surgery, Chengdu Anorectal Hospital, Chengdu, China
| | - Tianbao Xiao
- Colorectal and Anal Surgery, the First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
8
|
Tatum NJ, Endicott JA. Chatterboxes: the structural and functional diversity of cyclins. Semin Cell Dev Biol 2020; 107:4-20. [PMID: 32414682 DOI: 10.1016/j.semcdb.2020.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/16/2022]
Abstract
Proteins of the cyclin family have divergent sequences and execute diverse roles within the cell while sharing a common fold: the cyclin box domain. Structural studies of cyclins have played a key role in our characterization and understanding of cellular processes that they control, though to date only ten of the 29 CDK-activating cyclins have been structurally characterized by X-ray crystallography or cryo-electron microscopy with or without their cognate kinases. In this review, we survey the available structures of human cyclins, highlighting their molecular features in the context of their cellular roles. We pay particular attention to how cyclin activity is regulated through fine control of degradation motif recognition and ubiquitination. Finally, we discuss the emergent roles of cyclins independent of their roles as cyclin-dependent protein kinase activators, demonstrating the cyclin box domain to be a versatile and generalized scaffolding domain for protein-protein interactions across the cellular machinery.
Collapse
Affiliation(s)
- Natalie J Tatum
- Cancer Research UK Newcastle Drug Discovery Unit, Newcastle Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Jane A Endicott
- Cancer Research UK Newcastle Drug Discovery Unit, Newcastle Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom.
| |
Collapse
|
9
|
Li Y, Guo H, Wang Z, Bu H, Wang S, Wang H, Fang H, Liu Z, Kong B. Cyclin F and KIF20A, FOXM1 target genes, increase proliferation and invasion of ovarian cancer cells. Exp Cell Res 2020; 395:112212. [PMID: 32771525 DOI: 10.1016/j.yexcr.2020.112212] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/17/2020] [Accepted: 07/30/2020] [Indexed: 01/11/2023]
Abstract
Increased expression of FOXM1 is observed in a variety of human malignancies. The downstream target genes of FOXM1 involved in tumorigenesis and development are not fully elucidated in ovarian cancer. Here, we identified Cyclin F, a substrate recognition subunit of SCF (Skp1-Cul1-F-box protein) complex, and Kinesin Family Member 20A (KIF20A) were transcriptionally regulated by FOXM1 in ovarian cancer. Accordingly, Cyclin F and KIF20A were commonly overexpressed in ovarian cancer. Functionally, forced expression of Cyclin F or KIF20A significantly enhanced while knockdown of them decreased proliferation and invasion of ovarian cancer cells. Importantly, high levels of Cyclin F and KIF20A correlated with poor prognosis in patients with ovarian cancer. Our findings indicate that Cyclin F and KIF20A are functional targets of FOXM1 which might be potential drug targets in ovarian cancer.
Collapse
Affiliation(s)
- Yingwei Li
- School of Medicine, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, 250012, China
| | - Haiyang Guo
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Ji'nan, China
| | - Zixiang Wang
- School of Medicine, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, 250012, China
| | - Hualei Bu
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shourong Wang
- School of Medicine, Cheeloo College of Medicine, Shandong University, Ji'nan, Shandong, 250012, China
| | - Hao Wang
- Advanced Medical Research Institute, Shandong University, Jinan, 250012, China
| | - Haiyan Fang
- Department of Obstetrics & Gynecology, Jinhua Hospital of Zhejiang University, Jinhua, 321000, China
| | - Zhaojian Liu
- Department of Cell Biology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
10
|
Fulcher LJ, Sapkota GP. Mitotic kinase anchoring proteins: the navigators of cell division. Cell Cycle 2020; 19:505-524. [PMID: 32048898 PMCID: PMC7100989 DOI: 10.1080/15384101.2020.1728014] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/14/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
The coordinated activities of many protein kinases, acting on multiple protein substrates, ensures the error-free progression through mitosis of eukaryotic cells. Enormous research effort has thus been devoted to studying the roles and regulation of these mitotic kinases, and to the identification of their physiological substrates. Central for the timely deployment of specific protein kinases to their appropriate substrates during the cell division cycle are the many anchoring proteins, which serve critical regulatory roles. Through direct association, anchoring proteins are capable of modulating the catalytic activity and/or sub-cellular distribution of the mitotic kinases they associate with. The key roles of some anchoring proteins in cell division are well-established, whilst others are still being unearthed. Here, we review the current knowledge on anchoring proteins for some mitotic kinases, and highlight how targeting anchoring proteins for inhibition, instead of the mitotic kinases themselves, could be advantageous for disrupting the cell division cycle.
Collapse
Affiliation(s)
- Luke J Fulcher
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Scotland, UK
| | - Gopal P Sapkota
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|
11
|
Zaveri L, Dhawan J. Cycling to Meet Fate: Connecting Pluripotency to the Cell Cycle. Front Cell Dev Biol 2018; 6:57. [PMID: 29974052 PMCID: PMC6020794 DOI: 10.3389/fcell.2018.00057] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 05/14/2018] [Indexed: 01/26/2023] Open
Abstract
Pluripotent stem cells are characterized by their high proliferative rates, their ability to self-renew and their potential to differentiate to all the three germ layers. This rapid proliferation is brought about by a highly modified cell cycle that allows the cells to quickly shuttle from DNA synthesis to cell division, by reducing the time spent in the intervening gap phases. Many key regulators that define the somatic cell cycle are either absent or exhibit altered behavior, allowing the pluripotent cell to bypass cell cycle checkpoints typical of somatic cells. Experimental analysis of this modified stem cell cycle has been challenging due to the strong link between rapid proliferation and pluripotency, since perturbations to the cell cycle or pluripotency factors result in differentiation. Despite these hurdles, our understanding of this unique cell cycle has greatly improved over the past decade, in part because of the availability of new technologies that permit the analysis of single cells in heterogeneous populations. This review aims to highlight some of the recent discoveries in this area with a special emphasis on different states of pluripotency. We also discuss the highly interlinked network that connects pluripotency factors and key cell cycle genes and review evidence for how this interdependency may promote the rapid cell cycle. This issue gains translational importance since disruptions in stem cell proliferation and differentiation can impact disorders at opposite ends of a spectrum, from cancer to degenerative disease.
Collapse
Affiliation(s)
- Lamuk Zaveri
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India.,Manipal Academy of Higher Education, Manipal, India
| | - Jyotsna Dhawan
- Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India.,CSIR - Centre for Cellular and Molecular Biology, Hyderabad, India
| |
Collapse
|
12
|
Hassani S, Khaleghian A, Ahmadian S, Alizadeh S, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. Redistribution of cell cycle by arsenic trioxide is associated with demethylation and expression changes of cell cycle related genes in acute promyelocytic leukemia cell line (NB4). Ann Hematol 2017; 97:83-93. [DOI: 10.1007/s00277-017-3163-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 10/24/2017] [Indexed: 01/11/2023]
|
13
|
Alghamian Y, Abou Alchamat G, Murad H, Madania A. Effects of γ-radiation on cell growth, cell cycle and promoter methylation of 22 cell cycle genes in the 1321NI astrocytoma cell line. Adv Med Sci 2017; 62:330-337. [PMID: 28511071 DOI: 10.1016/j.advms.2017.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2016] [Revised: 03/02/2017] [Accepted: 03/09/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE DNA damage caused by radiation initiates biological responses affecting cell fate. DNA methylation regulates gene expression and modulates DNA damage pathways. Alterations in the methylation profiles of cell cycle regulating genes may control cell response to radiation. In this study we investigated the effect of ionizing radiation on the methylation levels of 22 cell cycle regulating genes in correlation with gene expression in 1321NI astrocytoma cell line. METHODS 1321NI cells were irradiated with 2, 5 or 10Gy doses then analyzed after 24, 48 and 72h for cell viability using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliu bromide) assay. Flow cytometry were used to study the effect of 10Gy irradiation on cell cycle. EpiTect Methyl II PCR Array was used to identify differentially methylated genes in irradiated cells. Changes in gene expression was determined by qPCR. Azacytidine treatment was used to determine whether DNA methylation affectes gene expression. RESULTS Our results showed that irradiation decreased cell viability and caused cell cycle arrest at G2/M. Out of 22 genes tested, only CCNF and RAD9A showed some increase in DNA methylation (3.59% and 3.62%, respectively) after 10Gy irradiation, and this increase coincided with downregulation of both genes (by 4 and 2 fold, respectively). TREATMENT with azacytidine confirmed that expression of CCNF and RAD9A genes was regulated by methylation. CONCLUSIONS 1321NI cell line is highly radioresistant and that irradiation of these cells with a 10Gy dose increases DNA methylation of CCNF and RAD9A genes. This dose down-regulates these genes, favoring G2/M arrest.
Collapse
|
14
|
Galper J, Rayner SL, Hogan AL, Fifita JA, Lee A, Chung RS, Blair IP, Yang S. Cyclin F: A component of an E3 ubiquitin ligase complex with roles in neurodegeneration and cancer. Int J Biochem Cell Biol 2017; 89:216-220. [DOI: 10.1016/j.biocel.2017.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/05/2017] [Accepted: 06/22/2017] [Indexed: 12/13/2022]
|
15
|
Furqan MS, Siyal MY. Elastic-Net Copula Granger Causality for Inference of Biological Networks. PLoS One 2016; 11:e0165612. [PMID: 27792750 PMCID: PMC5085021 DOI: 10.1371/journal.pone.0165612] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 10/15/2016] [Indexed: 12/13/2022] Open
Abstract
AIM In bioinformatics, the inference of biological networks is one of the most active research areas. It involves decoding various complex biological networks that are responsible for performing diverse functions in human body. Among these networks analysis, most of the research focus is towards understanding effective brain connectivity and gene networks in order to cure and prevent related diseases like Alzheimer and cancer respectively. However, with recent advances in data procurement technology, such as DNA microarray analysis and fMRI that can simultaneously process a large amount of data, it yields high-dimensional data sets. These high dimensional dataset analyses possess challenges for the analyst. BACKGROUND Traditional methods of Granger causality inference use ordinary least-squares methods for structure estimation, which confront dimensionality issues when applied to high-dimensional data. Apart from dimensionality issues, most existing methods were designed to capture only the linear inferences from time series data. METHOD AND CONCLUSION In this paper, we address the issues involved in assessing Granger causality for both linear and nonlinear high-dimensional data by proposing an elegant form of the existing LASSO-based method that we call "Elastic-Net Copula Granger causality". This method provides a more stable way to infer biological networks which has been verified using rigorous experimentation. We have compared the proposed method with the existing method and demonstrated that this new strategy outperforms the existing method on all measures: precision, false detection rate, recall, and F1 score. We have also applied both methods to real HeLa cell data and StarPlus fMRI datasets and presented a comparison of the effectiveness of both methods.
Collapse
Affiliation(s)
- Mohammad Shaheryar Furqan
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore, Singapore
- INFINITUS, Infocomm Centre of Excellence, Nanyang Technological University, Singapore, Singapore
| | - Mohammad Yakoob Siyal
- School of Electrical and Electronic Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
16
|
Soares JR, José Tenório de Melo E, da Cunha M, Fernandes KVS, Taveira GB, da Silva Pereira L, Pimenta S, Trindade FG, Regente M, Pinedo M, de la Canal L, Gomes VM, de Oliveira Carvalho A. Interaction between the plant ApDef 1 defensin and Saccharomyces cerevisiae results in yeast death through a cell cycle- and caspase-dependent process occurring via uncontrolled oxidative stress. Biochim Biophys Acta Gen Subj 2016; 1861:3429-3443. [PMID: 27614033 DOI: 10.1016/j.bbagen.2016.09.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 08/24/2016] [Accepted: 09/04/2016] [Indexed: 01/15/2023]
Abstract
BACKGROUND Plant defensins were discovered at beginning of the 90s'; however, their precise mechanism of action is still unknown. Herein, we studied ApDef1-Saccharomyces cerevisiae interaction. METHODS ApDef1-S. cerevisiae interaction was studied by determining the MIC, viability and death kinetic assays. Viability assay was repeated with hydroxyurea synchronized-yeast and pretreated with CCCP. Plasma membrane permeabilization, ROS induction, chromatin condensation, and caspase activation analyses were assessed through Sytox green, DAB, DAPI and FITC-VAD-FMK, respectively. Viability assay was done in presence of ascorbic acid and Z-VAD-FMK. Ultrastructural analysis was done by electron microscopy. RESULTS ApDef1 caused S. cerevisiae cell death and MIC was 7.8μM. Whole cell population died after 18h of ApDef1 interaction. After 3h, 98.76% of synchronized cell population died. Pretreatment with CCCP protected yeast from ApDef1 induced death. ApDef1-S. cerevisiae interaction resulted in membrane permeabilization, H2O2 increased production, chromatin condensation and caspase activation. Ascorbic acid prevented yeast cell death and membrane permeabilization. Z-VAD-FMK prevented yeast cell death. CONCLUSIONS ApDef1-S. cerevisiae interaction caused cell death through cell cycle dependentprocess which requires preserved membrane potential. After interaction, yeast went through uncontrolled ROS production and accumulation, which led to plasma membrane permeabilization, chromatin condensation and, ultimately, cell death by activation of caspase-dependent apoptosis via. GENERAL SIGNIFICANCE We show novel requirements for the interaction between plant defensin and fungi cells, i.e. cell cycle phase and membrane potential, and we indicate that membrane permeabilization is probably caused by ROS and therefore, it would be an indirect event of the ApDef1-S. cerevisiae interaction.
Collapse
Affiliation(s)
- Júlia Ribeiro Soares
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - Edésio José Tenório de Melo
- Laboratório de Biologia Celular e Tecidual, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - Maura da Cunha
- Laboratório de Biologia Celular e Tecidual, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - Kátia Valevski Sales Fernandes
- Laboratório de Química e Função de Proteínas e Peptídeos, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - Gabriel Bonan Taveira
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - Lidia da Silva Pereira
- Laboratório de Melhoramento Genético Vegetal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - Samy Pimenta
- Laboratório de Melhoramento Genético Vegetal, Centro de Ciências e Tecnologias Agropecuárias, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - Fernanda Gomes Trindade
- Laboratório de Biologia Celular e Tecidual, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - Mariana Regente
- Instituto de Investigaciones Biologicas, Universidad Nacional de Mar del Plata -CONICET, Mar del Plata, Argentina
| | - Marcela Pinedo
- Instituto de Investigaciones Biologicas, Universidad Nacional de Mar del Plata -CONICET, Mar del Plata, Argentina
| | - Laura de la Canal
- Instituto de Investigaciones Biologicas, Universidad Nacional de Mar del Plata -CONICET, Mar del Plata, Argentina
| | - Valdirene Moreira Gomes
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil
| | - André de Oliveira Carvalho
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense - Darcy Ribeiro, Campos dos Goytacazes, RJ, Brazil.
| |
Collapse
|
17
|
Zheng N, Zhou Q, Wang Z, Wei W. Recent advances in SCF ubiquitin ligase complex: Clinical implications. Biochim Biophys Acta Rev Cancer 2016; 1866:12-22. [PMID: 27156687 DOI: 10.1016/j.bbcan.2016.05.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/03/2016] [Accepted: 05/04/2016] [Indexed: 12/09/2022]
Abstract
F-box proteins, which are subunit recruiting modules of SCF (SKP1-Cullin 1-F-box protein) E3 ligase complexes, play critical roles in the development and progression of human malignancies through governing multiple cellular processes including cell proliferation, apoptosis, invasion and metastasis. Moreover, there are emerging studies that lead to the development of F-box proteins inhibitors with promising therapeutic potential. In this article, we describe how F-box proteins including but not restricted to well-established Fbw7, Skp2 and β-TRCP, are involved in tumorigenesis. However, in-depth investigation is required to further explore the mechanism and the physiological contribution of undetermined F-box proteins in carcinogenesis. Lastly, we suggest that targeting F-box proteins could possibly open new avenues for the treatment and prevention of human cancers.
Collapse
Affiliation(s)
- Nana Zheng
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou 215123, China
| | - Quansheng Zhou
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou 215123, China
| | - Zhiwei Wang
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, the First Affiliated Hospital, Soochow University, Suzhou 215123, China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, MA 02215, USA.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, MA 02215, USA.
| |
Collapse
|
18
|
Zheng N, Wang Z, Wei W. Ubiquitination-mediated degradation of cell cycle-related proteins by F-box proteins. Int J Biochem Cell Biol 2016; 73:99-110. [PMID: 26860958 PMCID: PMC4798898 DOI: 10.1016/j.biocel.2016.02.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/05/2016] [Accepted: 02/05/2016] [Indexed: 02/06/2023]
Abstract
F-box proteins, subunits of SKP1-cullin 1-F-box protein (SCF) type of E3 ubiquitin ligase complexes, have been validated to play a crucial role in governing various cellular processes such as cell cycle, cell proliferation, apoptosis, migration, invasion and metastasis. Recently, a wealth of evidence has emerged that F-box proteins is critically involved in tumorigenesis in part through governing the ubiquitination and subsequent degradation of cell cycle proteins, and dysregulation of this process leads to aberrant cell cycle progression and ultimately, tumorigenesis. Therefore, in this review, we describe the critical role of F-box proteins in the timely regulation of cell cycle. Moreover, we discuss how F-box proteins involve in tumorigenesis via targeting cell cycle-related proteins using biochemistry studies, engineered mouse models, and pathological gene alternations. We conclude that inhibitors of F-box proteins could have promising therapeutic potentials in part through controlling of aberrant cell cycle progression for cancer therapies.
Collapse
Affiliation(s)
- Nana Zheng
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou 215123, China
| | - Zhiwei Wang
- The Cyrus Tang Hematology Center and Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, The First Affiliated Hospital, Soochow University, Suzhou 215123, China; Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA 02215, USA.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave., Boston, MA 02215, USA.
| |
Collapse
|
19
|
Vriens K, Cammue BPA, Thevissen K. Antifungal plant defensins: mechanisms of action and production. Molecules 2014; 19:12280-303. [PMID: 25153857 PMCID: PMC6271847 DOI: 10.3390/molecules190812280] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 07/29/2014] [Accepted: 08/04/2014] [Indexed: 12/18/2022] Open
Abstract
Plant defensins are small, cysteine-rich peptides that possess biological activity towards a broad range of organisms. Their activity is primarily directed against fungi, but bactericidal and insecticidal actions have also been reported. The mode of action of various antifungal plant defensins has been studied extensively during the last decades and several of their fungal targets have been identified to date. This review summarizes the mechanism of action of well-characterized antifungal plant defensins, including RsAFP2, MsDef1, MtDef4, NaD1 and Psd1, and points out the variety by which antifungal plant defensins affect microbial cell viability. Furthermore, this review summarizes production routes for plant defensins, either via heterologous expression or chemical synthesis. As plant defensins are generally considered non-toxic for plant and mammalian cells, they are regarded as attractive candidates for further development into novel antimicrobial agents.
Collapse
Affiliation(s)
- Kim Vriens
- Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, Kasteelpark Arenberg 20, Heverlee 3001, Belgium
| | - Bruno P A Cammue
- Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, Kasteelpark Arenberg 20, Heverlee 3001, Belgium.
| | - Karin Thevissen
- Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, Kasteelpark Arenberg 20, Heverlee 3001, Belgium
| |
Collapse
|
20
|
Lubovac-Pilav Z, Borràs DM, Ponce E, Louie MC. Using expression profiling to understand the effects of chronic cadmium exposure on MCF-7 breast cancer cells. PLoS One 2013; 8:e84646. [PMID: 24376830 PMCID: PMC3869932 DOI: 10.1371/journal.pone.0084646] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Accepted: 11/25/2013] [Indexed: 12/17/2022] Open
Abstract
Cadmium is a metalloestrogen known to activate the estrogen receptor and promote breast cancer cell growth. Previous studies have implicated cadmium in the development of more malignant tumors; however the molecular mechanisms behind this cadmium-induced malignancy remain elusive. Using clonal cell lines derived from exposing breast cancer cells to cadmium for over 6 months (MCF-7-Cd4, -Cd6, -Cd7, -Cd8 and -Cd12), this study aims to identify gene expression signatures associated with chronic cadmium exposure. Our results demonstrate that prolonged cadmium exposure does not merely result in the deregulation of genes but actually leads to a distinctive expression profile. The genes deregulated in cadmium-exposed cells are involved in multiple biological processes (i.e. cell growth, apoptosis, etc.) and molecular functions (i.e. cadmium/metal ion binding, transcription factor activity, etc.). Hierarchical clustering demonstrates that the five clonal cadmium cell lines share a common gene expression signature of breast cancer associated genes, clearly differentiating control cells from cadmium exposed cells. The results presented in this study offer insights into the cellular and molecular impacts of cadmium on breast cancer and emphasize the importance of studying chronic cadmium exposure as one possible mechanism of promoting breast cancer progression.
Collapse
Affiliation(s)
- Zelmina Lubovac-Pilav
- Systems Biology Research Centre – Bioinformatics, School of Bioscience, University of Skövde, Skövde, Sweden
- * E-mail: (MCL); (ZL)
| | - Daniel M. Borràs
- Systems Biology Research Centre – Bioinformatics, School of Bioscience, University of Skövde, Skövde, Sweden
| | - Esmeralda Ponce
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, California, United States of America
| | - Maggie C. Louie
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, California, United States of America
- College of Pharmacy, Touro University of California, Vallejo, California, United States of America
- * E-mail: (MCL); (ZL)
| |
Collapse
|
21
|
Nelson DE, Randle SJ, Laman H. Beyond ubiquitination: the atypical functions of Fbxo7 and other F-box proteins. Open Biol 2013; 3:130131. [PMID: 24107298 PMCID: PMC3814724 DOI: 10.1098/rsob.130131] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
F-box proteins (FBPs) are substrate-recruiting subunits of Skp1-cullin1-FBP (SCF)-type E3 ubiquitin ligases. To date, 69 FBPs have been identified in humans, but ubiquitinated substrates have only been identified for a few, with the majority of FBPs remaining ‘orphans’. In recent years, a growing body of work has identified non-canonical, SCF-independent roles for about 12% of the human FBPs. These atypical FBPs affect processes as diverse as transcription, cell cycle regulation, mitochondrial dynamics and intracellular trafficking. Here, we provide a general review of FBPs, with a particular emphasis on these expanded functions. We review Fbxo7 as an exemplar of this special group as it has well-defined roles in both SCF and non-SCF complexes. We review its function as a cell cycle regulator, via its ability to stabilize p27 protein and Cdk6 complexes, and as a proteasome regulator, owing to its high affinity binding to PI31. We also highlight recent advances in our understanding of Fbxo7 function in Parkinson's disease, where it functions in the regulation of mitophagy with PINK1 and Parkin. We postulate that a few extraordinary FBPs act as platforms that seamlessly segue their canonical and non-canonical functions to integrate different cellular pathways and link their regulation.
Collapse
Affiliation(s)
- David E Nelson
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, UK
| | | | | |
Collapse
|
22
|
Li L, Han ZY, Li CM, Jiang XQ, Wang GL. Upregulation of heat shock protein 32 in Sertoli cells alleviates the impairments caused by heat shock-induced apoptosis in mouse testis. Cell Stress Chaperones 2013; 18:333-51. [PMID: 23188493 PMCID: PMC3631093 DOI: 10.1007/s12192-012-0385-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 11/05/2012] [Accepted: 11/09/2012] [Indexed: 11/30/2022] Open
Abstract
Heat stress results in apoptosis in testicular germ cells. A small heat shock protein (hsp), hsp32, is induced by heat stress in the testis, but little is known about its definitive function in physiological processes. To clarify the underlying role of hsp32, hsp32 expression and related signals in the heat shock pathway were analysed in mouse testes and Sertoli cells after heat stress in vivo and in vitro; meanwhile, expression of hsp32 was silenced only in the Sertoli cells using three different small interfering RNAs (siRNAs) to further verify the functional role of hsp32 in Sertoli cells, and hsp32-derived carbon monoxide (CO) contents in cultured media were analysed to clarify whether hsp32-derived CO involve in the apoptosis regulation mechanisms. The results from the in vivo experiment showed that the high expression levels of hsp32 (P < 0.05) were observed whether chronic, moderate or acute, transient heat exposure. The in vitro experiment showed that acute, transient heat exposure resulted in increases in Sertoli cells apoptosis (P < 0.01), the expression of hsp32 and caspase-3 activity; hsp32-siRNA knockdown of hsp32 expression resulted in upregulated apoptosis (P < 0.01) and caspase-3 activity (P < 0.01) in the Sertoli cells and hyperthermia increases CO (P < 0.01) release by Sertoli cells. The results suggested that upregulating hsp32 in Sertoli cells inhibits caspase-3 activity and alleviates heat-induced impairments in mouse testis; hsp32-derived CO may involve in the regulation mechanism.
Collapse
Affiliation(s)
- Lian Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| | - Zhao-Yu Han
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| | - Cheng-Min Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| | - Xiao-Qiang Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| | - Gen-Lin Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| |
Collapse
|
23
|
Matz RL, Erickson B, Vaidyanathan S, Kukowska-Latallo JF, Baker JR, Orr BG, Banaszak Holl MM. Polyplex exposure inhibits cell cycle, increases inflammatory response, and can cause protein expression without cell division. Mol Pharm 2013; 10:1306-17. [PMID: 23458572 PMCID: PMC3637663 DOI: 10.1021/mp300470d] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We sought to evaluate the relationship between cell division and protein expression when using commercial poly(ethylenimine) (PEI)-based polyplexes. The membrane dye PKH26 was used to assess cell division, and cyan fluorescent protein (CFP) was used to monitor protein expression. When analyzed at the whole population level, a greater number of cells divided than expressed protein, regardless of the level of protein expression observed, giving apparent consistency with the hypothesis that protein expression requires cells to pass through mitosis in order for the transgene to overcome the nuclear membrane. However, when the polyplex-exposed population was evaluated for the amount of division in the protein-expressing subpopulation, it was observed that substantial amounts of expression had occurred in the absence of division. Indeed, in HeLa S3 cells, this represented the majority of expressing cells. Of interest, the doubling time for both cell lines was slowed by ~2-fold upon exposure to polyplexes. This change was not altered by the origin of the plasmid DNA (pDNA) transgene promoter (cytomegalovirus (CMV) or elongation factor-1 alpha (EF1α)). Gene expression arrays in polyplex-exposed HeLa S3 cells showed upregulation of cell cycle arrest genes and downregulation of genes related to mitosis. Chemokine, interleukin, and toll-like receptor genes were also upregulated, suggesting activation of proinflammatory pathways. In summary, we find evidence that a cell division-independent expression pathway exists, and that polyplex exposure slows cell division and increases inflammatory response.
Collapse
Affiliation(s)
- Rebecca L. Matz
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109
| | - Blake Erickson
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109
- Program in Biophysics, University of Michigan, Ann Arbor, MI 48109
| | - Sriram Vaidyanathan
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Jolanta F. Kukowska-Latallo
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - James R. Baker
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| | - Bradford G. Orr
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109
- Department of Physics, University of Michigan, Ann Arbor, MI 48109
| | - Mark M. Banaszak Holl
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109
- Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109
- Program in Biophysics, University of Michigan, Ann Arbor, MI 48109
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
24
|
Yap TA, Molife LR, Blagden SP, de Bono S. Targeting cell cycle kinases and kinesins in anticancer drug development. Expert Opin Drug Discov 2013; 2:539-60. [PMID: 23484760 DOI: 10.1517/17460441.2.4.539] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The cell cycle is regulated by kinases such as the cyclin-dependent kinases (CDKs) and non-CDKs, which include Aurora and polo-like kinases, as well as checkpoint proteins. Mitotic kinesins are involved in the establishment of the mitotic spindle formation and function, and also play a role in cell cycle control. The disruption of the cell cycle is a hallmark of malignancy. Genetic or epigenetic events result in the upregulation of these kinases and mitotic kinesins in a myriad of tumour types, suggesting that their inhibition could result in preferential targeting of malignant cells. Such findings make the development of these inhibitors a rational and attractive new area for cancer therapeutics. Although challenges of potency and non-specificity have hampered their progress through the clinic, several novel compounds are presently in various phases of clinical trial evaluation.
Collapse
Affiliation(s)
- Timothy A Yap
- Drug Development Unit, The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey, SM2 5PT, UK.
| | | | | | | |
Collapse
|
25
|
Mishra R. Cell cycle-regulatory cyclins and their deregulation in oral cancer. Oral Oncol 2013; 49:475-81. [PMID: 23434055 DOI: 10.1016/j.oraloncology.2013.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 01/24/2013] [Accepted: 01/25/2013] [Indexed: 11/26/2022]
Abstract
Oral cancer is a growth-related disorder, and cyclins are the prime regulators of cell division. Cyclins are associated with the pathogenesis of oral cancer and are considered valuable biomarkers for diagnosis and prognosis. These important molecules are regulated in many ways to achieve a gain in function and are involved in promoting neoplastic growth. While the causes of most cyclin overexpression are varied, these cyclins may be induced by buccal mucosal insult mainly with carcinogens that alter various pathways propelling oral cancer. Substantial experimental evidences support a link between oncogenic signaling pathways and the deregulation of cyclins in oral cancer. This review focuses on the mechanisms by which cyclins are regulated and promote oral oncogenesis.
Collapse
Affiliation(s)
- Rajakishore Mishra
- Centre for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ratu-Lohardaga Road, Brambe, Ranchi 835 205, Jharkhand, India.
| |
Collapse
|
26
|
Fu J, Qiu H, Cai M, Pan Y, Cao Y, Liu L, Yun J, Zhang CZ. Low cyclin F expression in hepatocellular carcinoma associates with poor differentiation and unfavorable prognosis. Cancer Sci 2013; 104:508-15. [PMID: 23305207 DOI: 10.1111/cas.12100] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 12/25/2012] [Accepted: 01/06/2013] [Indexed: 02/07/2023] Open
Abstract
Cyclin F, capable of forming Skp1-Cul1-F-box protein ubiquitin ligase complex, is implicated in controlling centrosome duplication and preventing genome instability. Cyclin F oscillates during cell cycle with a similar pattern to cyclin A. However, its expression and significance in cancer remain obscure. In this study, we showed that cyclin F was noticeably decreased in 16 pairs of tissue samples of hepatocellular carcinoma (HCC) compared to paracarcinoma tissues, at both mRNA and protein levels. Immunohistochemical staining data revealed that in 71.8% (176/245) of HCC cases, cyclin F expression in tumor tissue was much lower than that in nontumorous tissue. Low cyclin F expression, defined by receiver operating characteristic curve analysis, was present in 69.0% of HCC patients. Low expression of cyclin F was significantly correlated with tumor size, clinical stage, serum alpha-fetoprotein level and tumor multiplicity. Further study showed that cyclin F expression was reversely associated with tumor differentiation in HCC. Kaplan-Meier analysis indicated that low cyclin F expression was related to poor overall survival and recurrence-free survival. The prognostic impact of cyclin F was further confirmed by stratified survival analysis. Importantly, multivariate analysis revealed that low cyclin F expression was an independent poor prognostic marker for overall survival. We conclude that cyclin F is downregulated in HCC and is a promising prognostic marker for patients suffering from this deadly disease.
Collapse
Affiliation(s)
- Jia Fu
- State Key Laboratory of Oncology in South China, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
27
|
D'Angiolella V, Esencay M, Pagano M. A cyclin without cyclin-dependent kinases: cyclin F controls genome stability through ubiquitin-mediated proteolysis. Trends Cell Biol 2012. [PMID: 23182110 DOI: 10.1016/j.tcb.2012.10.011] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cell cycle transitions are driven by the periodic oscillations of cyclins, which bind and activate cyclin-dependent kinases (CDKs) to phosphorylate target substrates. Cyclin F uses a substrate recruitment strategy similar to that of the other cyclins, but its associated catalytic activity is substantially different. Indeed, cyclin F is the founding member of the F-box family of proteins, which are the substrate recognition subunits of Skp1-Cul1-F-box protein (SCF) ubiquitin ligase complexes. Here, we discuss cyclin F function and recently identified substrates of SCF(cyclin)(F) involved in deoxyribonucleotide triphosphate (dNTP) production, centrosome duplication, and spindle formation. We highlight the relevance of cyclin F in controlling genome stability through ubiquitin-mediated proteolysis and the implications for cancer development.
Collapse
Affiliation(s)
- Vincenzo D'Angiolella
- Department of Pathology, NYU Cancer Institute, New York University School of Medicine, New York, NY 10016, USA. vincenzo.d'
| | | | | |
Collapse
|
28
|
Novel mithramycins abrogate the involvement of protein factors in the transcription of cell cycle control genes. Biochem Pharmacol 2012; 84:1133-42. [DOI: 10.1016/j.bcp.2012.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/20/2012] [Accepted: 08/03/2012] [Indexed: 12/11/2022]
|
29
|
Benisch P, Schilling T, Klein-Hitpass L, Frey SP, Seefried L, Raaijmakers N, Krug M, Regensburger M, Zeck S, Schinke T, Amling M, Ebert R, Jakob F. The transcriptional profile of mesenchymal stem cell populations in primary osteoporosis is distinct and shows overexpression of osteogenic inhibitors. PLoS One 2012; 7:e45142. [PMID: 23028809 PMCID: PMC3454401 DOI: 10.1371/journal.pone.0045142] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 08/13/2012] [Indexed: 12/11/2022] Open
Abstract
Primary osteoporosis is an age-related disease characterized by an imbalance in bone homeostasis. While the resorptive aspect of the disease has been studied intensely, less is known about the anabolic part of the syndrome or presumptive deficiencies in bone regeneration. Multipotent mesenchymal stem cells (MSC) are the primary source of osteogenic regeneration. In the present study we aimed to unravel whether MSC biology is directly involved in the pathophysiology of the disease and therefore performed microarray analyses of hMSC of elderly patients (79–94 years old) suffering from osteoporosis (hMSC-OP). In comparison to age-matched controls we detected profound changes in the transcriptome in hMSC-OP, e.g. enhanced mRNA expression of known osteoporosis-associated genes (LRP5, RUNX2, COL1A1) and of genes involved in osteoclastogenesis (CSF1, PTH1R), but most notably of genes coding for inhibitors of WNT and BMP signaling, such as Sclerostin and MAB21L2. These candidate genes indicate intrinsic deficiencies in self-renewal and differentiation potential in osteoporotic stem cells. We also compared both hMSC-OP and non-osteoporotic hMSC-old of elderly donors to hMSC of ∼30 years younger donors and found that the transcriptional changes acquired between the sixth and the ninth decade of life differed widely between osteoporotic and non-osteoporotic stem cells. In addition, we compared the osteoporotic transcriptome to long term-cultivated, senescent hMSC and detected some signs for pre-senescence in hMSC-OP. Our results suggest that in primary osteoporosis the transcriptomes of hMSC populations show distinct signatures and little overlap with non-osteoporotic aging, although we detected some hints for senescence-associated changes. While there are remarkable inter-individual variations as expected for polygenetic diseases, we could identify many susceptibility genes for osteoporosis known from genetic studies. We also found new candidates, e.g. MAB21L2, a novel repressor of BMP-induced transcription. Such transcriptional changes may reflect epigenetic changes, which are part of a specific osteoporosis-associated aging process.
Collapse
Affiliation(s)
- Peggy Benisch
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Tatjana Schilling
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Ludger Klein-Hitpass
- Institute of Cell Biology (Tumor Research), University Hospital Essen, Essen, Germany
| | - Sönke P. Frey
- Department of Trauma, Hand-, Plastic- and Reconstructive Surgery, University Hospital of Wuerzburg, Wuerzburg, Germany
| | - Lothar Seefried
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Nadja Raaijmakers
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Melanie Krug
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Martina Regensburger
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Sabine Zeck
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Regina Ebert
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
| | - Franz Jakob
- Orthopedic Center for Musculoskeletal Research, University of Wuerzburg, Wuerzburg, Germany
- * E-mail:
| |
Collapse
|
30
|
A p27Kip1 mutant that does not inhibit CDK activity promotes centrosome amplification and micronucleation. Oncogene 2011; 31:3989-98. [PMID: 22158041 PMCID: PMC3432229 DOI: 10.1038/onc.2011.550] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Mitotic catastrophe occurs when cells enter mitosis with damaged DNA or excess centrosomes. Cells overexpressing the centrosome protein CP110 or depleted of cyclin F, which targets CP110 for destruction, have more than two centrosomes and undergo mitotic catastrophe. Our studies show centrosome reduplication and mitotic catastrophe in osteosarcoma cells inducibly expressing a p27Kip1 mutant (termed p27K) that binds cyclins but not cyclin-dependent kinases (CDKs). p27K inhibited cell proliferation but not CDK activity or cell cycle progression. It did not induce apoptosis; however, cells expressing p27K had more than two centrosomes and, indicative of mitotic catastrophe, irregularly shaped nuclei or multiple micronuclei. p27K interacted with cyclin F in vivo (as did endogenous p27Kip1) and displaced cyclin F from CP110. Depletion of CP110 rescued p27K-expressing cells from centrosome reduplication and mitotic catastrophe. Collectively, our data show that p27Kip1 can perturb mitosis and suggest that it does so by sequestering cyclin F, which prevents its interaction with and the subsequent degradation of CP110, ultimately resulting in centrosome reduplication, mitotic catastrophe and abrogation of cell proliferation.
Collapse
|
31
|
Constraint-based analysis of gene interactions using restricted boolean networks and time-series data. BMC Proc 2011; 5 Suppl 2:S5. [PMID: 21554763 PMCID: PMC3090763 DOI: 10.1186/1753-6561-5-s2-s5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background A popular model for gene regulatory networks is the Boolean network model. In this paper, we propose an algorithm to perform an analysis of gene regulatory interactions using the Boolean network model and time-series data. Actually, the Boolean network is restricted in the sense that only a subset of all possible Boolean functions are considered. We explore some mathematical properties of the restricted Boolean networks in order to avoid the full search approach. The problem is modeled as a Constraint Satisfaction Problem (CSP) and CSP techniques are used to solve it. Results We applied the proposed algorithm in two data sets. First, we used an artificial dataset obtained from a model for the budding yeast cell cycle. The second data set is derived from experiments performed using HeLa cells. The results show that some interactions can be fully or, at least, partially determined under the Boolean model considered. Conclusions The algorithm proposed can be used as a first step for detection of gene/protein interactions. It is able to infer gene relationships from time-series data of gene expression, and this inference process can be aided by a priori knowledge available.
Collapse
|
32
|
Iwamoto K, Hamada H, Eguchi Y, Okamoto M. Mathematical modeling of cell cycle regulation in response to DNA damage: Exploring mechanisms of cell-fate determination. Biosystems 2011; 103:384-91. [DOI: 10.1016/j.biosystems.2010.11.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 11/09/2010] [Accepted: 11/15/2010] [Indexed: 10/18/2022]
|
33
|
Drafahl KA, McAndrew CW, Meyer AN, Haas M, Donoghue DJ. The receptor tyrosine kinase FGFR4 negatively regulates NF-kappaB signaling. PLoS One 2010; 5:e14412. [PMID: 21203561 PMCID: PMC3008709 DOI: 10.1371/journal.pone.0014412] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 11/24/2010] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND NFκB signaling is of paramount importance in the regulation of apoptosis, proliferation, and inflammatory responses during human development and homeostasis, as well as in many human cancers. Receptor Tyrosine Kinases (RTKs), including the Fibroblast Growth Factor Receptors (FGFRs) are also important in development and disease. However, a direct relationship between growth factor signaling pathways and NFκB activation has not been previously described, although FGFs have been known to antagonize TNFα-induced apoptosis. METHODOLOGY/PRINCIPAL FINDINGS Here, we demonstrate an interaction between FGFR4 and IKKβ (Inhibitor of NFκB Kinase β subunit), an essential component in the NFκB pathway. This novel interaction was identified utilizing a yeast two-hybrid screen [1] and confirmed by coimmunoprecipitation and mass spectrometry analysis. We demonstrate tyrosine phosphorylation of IKKβ in the presence of activated FGFR4, but not kinase-dead FGFR4. Following stimulation by TNFα (Tumor Necrosis Factor α) to activate NFκB pathways, FGFR4 activation results in significant inhibition of NFκB signaling as measured by decreased nuclear NFκB localization, by reduced NFκB transcriptional activation in electophoretic mobility shift assays, and by inhibition of IKKβ kinase activity towards the substrate GST-IκBα in in vitro assays. FGF19 stimulation of endogenous FGFR4 in TNFα-treated DU145 prostate cancer cells also leads to a decrease in IKKβ activity, concomitant reduction in NFκB nuclear localization, and reduced apoptosis. Microarray analysis demonstrates that FGF19 + TNFα treatment of DU145 cells, in comparison with TNFα alone, favors proliferative genes while downregulating genes involved in apoptotic responses and NFκB signaling. CONCLUSIONS/SIGNIFICANCE These results identify a compelling link between FGFR4 signaling and the NFκB pathway, and reveal that FGFR4 activation leads to a negative effect on NFκB signaling including an inhibitory effect on proapoptotic signaling. We anticipate that this interaction between an RTK and a component of NFκB signaling will not be limited to FGFR4 alone.
Collapse
Affiliation(s)
- Kristine A. Drafahl
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Christopher W. McAndrew
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - April N. Meyer
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
| | - Martin Haas
- Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
| | - Daniel J. Donoghue
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California, United States of America
- Moores Cancer Center, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Deshpande R, Sharma S, Verfaillie CM, Hu WS, Myers CL. A scalable approach for discovering conserved active subnetworks across species. PLoS Comput Biol 2010; 6:e1001028. [PMID: 21170309 PMCID: PMC3000367 DOI: 10.1371/journal.pcbi.1001028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Accepted: 11/10/2010] [Indexed: 12/31/2022] Open
Abstract
Overlaying differential changes in gene expression on protein interaction networks has proven to be a useful approach to interpreting the cell's dynamic response to a changing environment. Despite successes in finding active subnetworks in the context of a single species, the idea of overlaying lists of differentially expressed genes on networks has not yet been extended to support the analysis of multiple species' interaction networks. To address this problem, we designed a scalable, cross-species network search algorithm, neXus (Network - cross(X)-species - Search), that discovers conserved, active subnetworks based on parallel differential expression studies in multiple species. Our approach leverages functional linkage networks, which provide more comprehensive coverage of functional relationships than physical interaction networks by combining heterogeneous types of genomic data. We applied our cross-species approach to identify conserved modules that are differentially active in stem cells relative to differentiated cells based on parallel gene expression studies and functional linkage networks from mouse and human. We find hundreds of conserved active subnetworks enriched for stem cell-associated functions such as cell cycle, DNA repair, and chromatin modification processes. Using a variation of this approach, we also find a number of species-specific networks, which likely reflect mechanisms of stem cell function that have diverged between mouse and human. We assess the statistical significance of the subnetworks by comparing them with subnetworks discovered on random permutations of the differential expression data. We also describe several case examples that illustrate the utility of comparative analysis of active subnetworks. Microarrays are a powerful tool for discovering genes whose expression is associated with a particular biological process or phenotype. Differential expression analysis can often generate a list of several hundred or even thousands of significant genes. While these genes represent real expression differences, the large number of candidates can make the process of hypothesis generation for further experimental studies challenging. Use of complementary datasets such as protein-protein interactions can help filter such candidate lists to genes involved with the most relevant pathways. This approach has been applied successfully by many groups, but to date, no one has developed an approach for discovering active pathways or subnetworks that are conserved across multiple species. We propose an algorithm, neXus (Network – cross(X)-species – Search), for cross-species active subnetwork discovery given candidate gene lists from two species and weighted protein-protein interaction networks. We validate our approach on expression studies from human and mouse stem cells. We find many active subnetworks that are conserved across species relevant to stem cell biology as well as other subnetworks that show species-specific behavior. We show that these networks are not likely to have been discovered by chance and discuss several specific cases that reveal potentially novel stem cell biology.
Collapse
Affiliation(s)
- Raamesh Deshpande
- Department of Computer Science and Engineering, University of Minnesota - Twin Cities, Minneapolis, Minnesota, United States of America
| | - Shikha Sharma
- Department of Chemical Engineering, University of Minnesota - Twin Cities, Minneapolis, Minnesota, United States of America
| | | | - Wei-Shou Hu
- Department of Chemical Engineering, University of Minnesota - Twin Cities, Minneapolis, Minnesota, United States of America
| | - Chad L. Myers
- Department of Computer Science and Engineering, University of Minnesota - Twin Cities, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
35
|
Hayano T, Koiwai K, Ishii H, Maezawa S, Kouda K, Motoyama T, Kubota T, Koiwai O. TdT interacting factor 1 enhances TdT ubiquitylation through recruitment of BPOZ-2 into nucleus from cytoplasm. Genes Cells 2009; 14:1415-27. [PMID: 19930467 DOI: 10.1111/j.1365-2443.2009.01358.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Takahide Hayano
- Faculty of Science & Technology, Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Hancock WW, Özkaynak E. Three distinct domains contribute to nuclear transport of murine Foxp3. PLoS One 2009; 4:e7890. [PMID: 19924293 PMCID: PMC2774276 DOI: 10.1371/journal.pone.0007890] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2009] [Accepted: 10/23/2009] [Indexed: 01/19/2023] Open
Abstract
Foxp3, a 47-kDa transcription factor, is necessary for the function of CD4+CD25+ regulatory T cells (Tregs), with an essential role in the control of self-reactive T cells and in preventing autoimmunity. Activation of Tregs by TCR engagement results in upregulation of Foxp3 expression, followed by its rapid nuclear transport and binding to chromatin. Here, we identify three distinct Foxp3 domains that contribute to nuclear transport. The first domain (Domain 1) comprises the C-terminal 12 amino acids. The second domain (Domain 2) is located immediately N-terminal to the forkhead domain (FHD), recently reported to be a binding site for the runt-related transcription factor 1/acute myeloid leukemia 1 (Runx1/AML1). The third domain (Domain 3) is located within the N-terminal first 51 amino acids. Unlike the known nuclear localization signals (NLSs), none of these three regions are rich in basic residues and do not bear any similarity to known monopartite or bipartite NLSs that have one or more clusters of basic amino acids. The basic arginine-lysine-lysine-arginine (RKKR) sequence, located 12-aa from the C-terminal end of Foxp3 was previously reported to be a nuclear localization signal (NLS) for several proteins, including for a GFP-Foxp3 hybrid. Evidence is provided here that in the full-length native Foxp3 RKKR does not function as an NLS. The data reported in this study indicates that Foxp3 achieves nuclear transport by binding to other nuclear factors and co-transporting with them to the nucleus.
Collapse
Affiliation(s)
- Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Engin Özkaynak
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
37
|
Carvalho ADO, Gomes VM. Plant defensins--prospects for the biological functions and biotechnological properties. Peptides 2009; 30:1007-20. [PMID: 19428780 DOI: 10.1016/j.peptides.2009.01.018] [Citation(s) in RCA: 168] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 01/22/2009] [Accepted: 01/26/2009] [Indexed: 01/07/2023]
Abstract
Plant defensins are a prominent family of cationic peptides in the plant kingdom. They are structurally and functionally related to defensins that have been previously characterized in mammals and insects. They present molecular masses between 5 and 7kDa and possess a pattern of eight conserved Cys residues. The three-dimensional structure of plant defensins is small and globular. It has three anti-parallel beta-sheets and one alpha-helix that is stabilized by a structural motif composed of disulfide bridges. This motif is found in other peptides with biological activity and is called the Cys stabilized alphabeta motif (CSalphabeta). Based on the growing knowledge on defensin structure, gene expression and regulation, and also their in vitro biological activity, it has become clear that plant defensins are complex and sophisticated peptides whose function extends beyond their role in defense of plants against microbial infection. This review discusses recent data and will present comprehensive information regarding the study of defensins.
Collapse
Affiliation(s)
- André de Oliveira Carvalho
- Laboratório de Fisiologia e Bioquímica de Microrganismos, Centro de Biociências e Biotecnologia, Universidade Estadual do Norte Fluminense-Darcy Ribeiro, Campos dos Goytacazes-RJ, Brazil.
| | | |
Collapse
|
38
|
Genoud T, Schweizer F, Tscheuschler A, Debrieux D, Casal JJ, Schäfer E, Hiltbrunner A, Fankhauser C. FHY1 mediates nuclear import of the light-activated phytochrome A photoreceptor. PLoS Genet 2008; 4:e1000143. [PMID: 18670649 PMCID: PMC2483295 DOI: 10.1371/journal.pgen.1000143] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Accepted: 06/25/2008] [Indexed: 01/30/2023] Open
Abstract
The phytochrome (phy) family of photoreceptors is of crucial importance throughout the life cycle of higher plants. Light-induced nuclear import is required for most phytochrome responses. Nuclear accumulation of phyA is dependent on two related proteins called FHY1 (Far-red elongated HYpocotyl 1) and FHL (FHY1 Like), with FHY1 playing the predominant function. The transcription of FHY1 and FHL are controlled by FHY3 (Far-red elongated HYpocotyl 3) and FAR1 (FAr-red impaired Response 1), a related pair of transcription factors, which thus indirectly control phyA nuclear accumulation. FHY1 and FHL preferentially interact with the light-activated form of phyA, but the mechanism by which they enable photoreceptor accumulation in the nucleus remains unsolved. Sequence comparison of numerous FHY1-related proteins indicates that only the NLS located at the N-terminus and the phyA-interaction domain located at the C-terminus are conserved. We demonstrate that these two parts of FHY1 are sufficient for FHY1 function. phyA nuclear accumulation is inhibited in the presence of high levels of FHY1 variants unable to enter the nucleus. Furthermore, nuclear accumulation of phyA becomes light- and FHY1-independent when an NLS sequence is fused to phyA, strongly suggesting that FHY1 mediates nuclear import of light-activated phyA. In accordance with this idea, FHY1 and FHY3 become functionally dispensable in seedlings expressing a constitutively nuclear version of phyA. Our data suggest that the mechanism uncovered in Arabidopsis is conserved in higher plants. Moreover, this mechanism allows us to propose a model explaining why phyA needs a specific nuclear import pathway.
Collapse
Affiliation(s)
- Thierry Genoud
- Centre for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Fabian Schweizer
- Centre for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Anke Tscheuschler
- Institut für Biologie II/Botanik, Albert Ludwigs Universität, Freiburg, Germany
| | - Dimitry Debrieux
- Centre for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Jorge J. Casal
- IFEVA, Facultad de Agronomía, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Eberhard Schäfer
- Institut für Biologie II/Botanik, Albert Ludwigs Universität, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Andreas Hiltbrunner
- Institut für Biologie II/Botanik, Albert Ludwigs Universität, Freiburg, Germany
- BIOSS, Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | | |
Collapse
|
39
|
Lu SY, Sontag DP, Detillieux KA, Cattini PA. FGF-16 is released from neonatal cardiac myocytes and alters growth-related signaling: a possible role in postnatal development. Am J Physiol Cell Physiol 2008; 294:C1242-9. [PMID: 18337564 DOI: 10.1152/ajpcell.00529.2007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
FGF-16 has been reported to be preferentially expressed in the adult rat heart. We have investigated the expression of FGF-16 in the perinatal and postnatal heart and its functional significance in neonatal rat cardiac myocytes. FGF-16 mRNA accumulation was observed by quantitative RT-PCR between neonatal days 1 and 7, with this increased expression persisting into adulthood. FGF-2 has been shown to increase neonatal rat cardiac myocyte proliferative potential via PKC activation. Gene array analysis revealed that FGF-16 inhibited the upregulation by FGF-2 of cell cycle promoting genes including cyclin F and Ki67. Furthermore, the CDK4/6 inhibitor gene Arf/INK4A was upregulated with the combination of FGF-16 and FGF-2 but not with either factor on its own. The effect on Ki67 was validated by protein immunodetection, which also showed that FGF-16 significantly decreased FGF-2-induced Ki67 labeling of cardiac myocytes, although it alone had no effect on Ki67 labeling. Inhibition of p38 MAPK potentiated cardiac myocyte proliferation induced by FGF-2 but did not alter the inhibitory action of FGF-16. Receptor binding assay showed that FGF-16 can compete with FGF-2 for binding sites including FGF receptor 1. FGF-16 had no effect on activated p38, ERK1/2, or JNK/SAPK after FGF-2 treatment. However, FGF-16 inhibited PKC-alpha and PKC-epsilon activation induced by FGF-2 and, importantly, IGF-1. Collectively, these data suggest that expression and release of FGF-16 in the neonatal myocardium interfere with cardiac myocyte proliferative potential by altering the local signaling environment via modulation of PKC activation and cell cycle-related gene expression.
Collapse
Affiliation(s)
- Shun Yan Lu
- Department of Physiology, University of Manitoba, Winnipeg, Manitoba, Canada R3E 3J7
| | | | | | | |
Collapse
|
40
|
Kaiserli E, Jenkins GI. UV-B promotes rapid nuclear translocation of the Arabidopsis UV-B specific signaling component UVR8 and activates its function in the nucleus. THE PLANT CELL 2007; 19:2662-73. [PMID: 17720867 PMCID: PMC2002606 DOI: 10.1105/tpc.107.053330] [Citation(s) in RCA: 190] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 07/13/2007] [Accepted: 08/09/2007] [Indexed: 05/16/2023]
Abstract
Arabidopsis thaliana UV RESISTANCE LOCUS8 (UVR8) is a UV-B-specific signaling component that binds to chromatin and regulates UV protection by orchestrating expression of a range of genes. Here, we studied how UV-B regulates UVR8. We show that UV-B stimulates the nuclear accumulation of both a green fluorescent protein (GFP)-UVR8 fusion and native UVR8. Nuclear accumulation leads to UV-B induction of the HY5 gene, encoding a key transcriptional effector of the UVR8 pathway. Nuclear accumulation of UVR8 is specific to UV-B, occurs at low fluence rates, and is observed within 5 min of UV-B exposure. Attachment of a nuclear export signal (NES) to GFP-UVR8 causes cytosolic localization in the absence of UV-B. However, UV-B promotes rapid nuclear accumulation of NES-GFP-UVR8, indicating a concerted mechanism for nuclear translocation. UVR8 lacking the N-terminal 23 amino acids is impaired in nuclear translocation. Attachment of a nuclear localization signal (NLS) to UVR8 causes constitutive nuclear localization. However, NLS-GFP-UVR8 only confers HY5 gene expression following UV-B illumination, indicating that nuclear localization, although necessary for UVR8 function, is insufficient to cause expression of target genes; UV-B is additionally required to stimulate UVR8 function in the nucleus. These findings provide new insights into the mechanisms through which UV-B regulates gene expression in plants.
Collapse
Affiliation(s)
- Eirini Kaiserli
- Plant Science Group, Division of Biochemistry and Molecular Biology, Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | | |
Collapse
|
41
|
Ersvaer E, Zhang JY, McCormack E, Olsnes A, Anensen N, Tan EM, Gjertsen BT, Bruserud O. Cyclin B1 is commonly expressed in the cytoplasm of primary human acute myelogenous leukemia cells and serves as a leukemia-associated antigen associated with autoantibody response in a subset of patients. Eur J Haematol 2007; 79:210-25. [PMID: 17655707 DOI: 10.1111/j.1600-0609.2007.00899.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Aberrant expression of cyclin B1, a cell cycle regulator, is related to prognosis in various human malignancies. Additionally, cytoplasmic expression of cyclin B1 in epithelial malignancies is associated with a specific T-cell response and presumably also a humoral immune response. We therefore investigated (i) whether a similar expression pattern could be detected in native human acute myelogenous leukemia (AML) cells and (ii) whether cyclin B1 specific antibodies could be detected in AML. METHODS AML cell expression of cyclin B1 was analyzed by flow cytometry and confocal microscopy. Humoral immune response in AML patient sera against cyclin B1 was analyzed by ELISA. RESULTS AML cell expression of cyclin B1 was detected for all 42 patients; but the percentage of cyclin B1 positive cells showed a wide variation between patients. Confocal microscopy demonstrated that 32/42 (76%) patient samples showed abnormal cytoplasmic expression. Furthermore, the cytoplasmic expression was maintained after 14 d of in vitro culture and differentiation of the AML cells towards a dendritic cell phenotype. Cyclin B1 specific serum antibodies could be detected for seven of 65 patients with untreated AML. CONCLUSIONS Our studies demonstrate that primary human AML cells show aberrant cytoplasmic expression of cyclin B1 for a majority of patients and a specific humoral immune response was also detected for a subset of patients with untreated leukemia.
Collapse
|
42
|
Bai J, Solberg C, Fernandes JMO, Johnston IA. Profiling of maternal and developmental-stage specific mRNA transcripts in Atlantic halibut Hippoglossus hippoglossus. Gene 2007; 386:202-10. [PMID: 17118579 DOI: 10.1016/j.gene.2006.09.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2005] [Revised: 09/04/2006] [Accepted: 09/19/2006] [Indexed: 11/22/2022]
Abstract
cDNA libraries were constructed from the following developmental stages (tissues) of the Atlantic halibut (Hippoglossus hippoglossus): 2-cell stage (embryos), 1 day-old yolk sac larvae (trunk) and juvenile (fast skeletal muscle). A total of 4249 high quality expressed sequence tags from the three libraries were clustered into a partial transcriptome of 2124 putative genes. A large proportion of the gene clusters (48.3%) had no significant matches against known proteins. The most abundant ESTs of nuclear transcripts in the 2-cell library included sequences with high identity to zebrafish H1M, a linker histone-like protein involved in primordial germ cell specification, zinc finger protein, rRNA external transcribed spacer, thymosin beta-4, cyclin B1 and several predicted peptides from the Tetraodon nigroviridis genome assembly with unknown functions. 170 and 123 ESTs represented ribosomal proteins in the larval and juvenile libraries respectively, compared with only two sequences in the 2-cell library, which may reflect an abundance of maternally inherited pre-formed ribosomes in the yolk. Even though some clusters were common to all three libraries, most putative genes showed a developmental-stage specific distribution with 72% (2-cell embryo), 59% (larval) and 57% (juvenile) sequences having no significant matches against the 8400 adult halibut sequences in the EMBL nucleotide database. Comparison between the predicted halibut peptide data set and the human, zebrafish, and pufferfishes (T. nigroviridis and Takifugu rubripes) proteomes revealed that, as expected, the halibut sequences were more similar to the other two fish species than to human proteins. However, no clear bias towards the pufferfishes was observed, suggesting significant sequence variation between orthologues within the clade Acanthomorpha. The sequence information generated in the present study will represent a significant new resource for future studies on normal and abnormal development in Atlantic halibut.
Collapse
Affiliation(s)
- Jialin Bai
- Fish Muscle Research Group, Gatty Marine Laboratory, School of Biology, University of St Andrews, East Sands, St Andrews, Fife, KY16 8LB, Scotland, UK
| | | | | | | |
Collapse
|
43
|
Hiltbrunner A, Viczián A, Bury E, Tscheuschler A, Kircher S, Tóth R, Honsberger A, Nagy F, Fankhauser C, Schäfer E. Nuclear accumulation of the phytochrome A photoreceptor requires FHY1. Curr Biol 2006; 15:2125-30. [PMID: 16332538 DOI: 10.1016/j.cub.2005.10.042] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Revised: 10/13/2005] [Accepted: 10/14/2005] [Indexed: 11/15/2022]
Abstract
The phytochrome family of red/far-red (R/FR)-responsive photoreceptors plays a key role throughout the life cycle of plants . Arabidopsis has five phytochromes, phyA-phyE, among which phyA and phyB play the most predominant functions . Light-regulated nuclear accumulation of the phytochromes is an important regulatory step of this pathway, but to this date no factor specifically required for this event has been identified . Among all phyA signaling mutants, fhy1 and fhy3 (far-red elongated hypocotyl 1 and 3) have the most severe hyposensitive phenotype, indicating that they play particularly important roles . FHY1 is a small plant-specific protein of unknown function localized both in the nucleus and the cytoplasm . Here we show that FHY1 is specifically required for the light-regulated nuclear accumulation of phyA but not phyB. Moreover, phyA accumulation is only slightly affected in fhy3, indicating that the diminished nuclear accumulation of phyA observed in fhy1 seedlings is not simply a general consequence of reduced phyA signaling. By in vitro pull-down and yeast two-hybrid analyses, we demonstrate that FHY1 physically interacts with phyA, preferentially in its active Pfr form. Furthermore, FHY1 and phyA colocalize in planta. We therefore identify the first component required for light-regulated phytochrome nuclear accumulation.
Collapse
Affiliation(s)
- Andreas Hiltbrunner
- Institut für Biologie II/Botanik, Albert Ludwigs Universität, Schänzlestrasse 1, D-79104 Freiburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Thermes V, Candal E, Alunni A, Serin G, Bourrat F, Joly JS. Medaka simplet (FAM53B) belongs to a family of novel vertebrate genes controlling cell proliferation. Development 2006; 133:1881-90. [PMID: 16611694 DOI: 10.1242/dev.02350] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The identification of genes that regulate proliferation is of great importance to developmental biology, regenerative medicine and cancer research. Using an in situ screen on a cortical structure of the medaka fish brain, we identified the simplet gene (smp), which is homologous to the human FAM53B gene. smp was expressed in actively proliferating cells of the CNS throughout embryogenesis. It belongs to a family of vertebrate-specific genes with no characterized biochemical domains. We showed that FAM53B bound 14-3-3 chaperones, as well as SKIIP proteins, adaptor proteins connecting DNA-binding proteins to modulators of transcription. smp inactivation with morpholinos led to delayed epiboly and reduced embryonic size. Absence of Smp activity did not induce apoptosis, but resulted in a reduced cell proliferation rate and enlarged blastomeres. Moreover, smp was shown to control the expression of the pluripotency-associated oct4/pou5f1 gene. We propose that smp is a novel vertebrate-specific gene needed for cell proliferation and that it is probably associated with the maintenance of a pluripotent state.
Collapse
Affiliation(s)
- Violette Thermes
- INRA MSNC Group, DEPSN, Institut A. Fessard, CNRS, 1 avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | | | | | | | | | | |
Collapse
|
45
|
Migone F, Deinnocentes P, Smith BF, Bird RC. Alterations in CDK1 expression and nuclear/nucleolar localization following induction in a spontaneous canine mammary cancer model. J Cell Biochem 2006; 98:504-18. [PMID: 16317763 DOI: 10.1002/jcb.20707] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Transcription of CDK1 is induced as cells re-enter the cell cycle from quiescence and these early cell cycle re-entry events have been modeled by okadaic acid treatment due to its activity on specific enhancer sequences in the human CDK1 promoter. To investigate heterogeneity of control of this mechanism in the context of neoplastic transformation, a cellular model derived from spontaneous canine mammary cancer (CMT) was developed that includes six cell lines derived from different animals. Notable heterogeneity in response to okadaic acid was observed in expression of CDK1 mRNA and protein. In response to okadaic acid treatment, two CMT cell lines exhibited a CDK1 mRNA induction while one cell line exhibited CDK1 mRNA suppression, and three remained unchanged. Despite this variability, three CMT cell lines arrested in S or G2/M phase and five exhibited marked increases in apoptosis. Moderation of some of these differences were observed at the level of CDK1 protein as three of six CMT cell lines exhibited only moderate enhancement in CDK1 protein levels while three remained essentially unchanged. Some additional differences in distribution of CDK1 protein, favoring enhanced nuclear over cytoplasmic CDK1 localization, were observed in treated cells in the form of concentrated nuclear CDK1 labeled foci. Confocal microscopy revealed the presence of brightly labeled punctate foci containing CDK1 protein within nuclei as well as nucleoli in okadaic acid treated non-mitotic cells suggesting a role for this kinase outside the normal G2/mitotic phase of the cell cycle and suggesting a possible new function within the nucleolus.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- CDC2 Protein Kinase/genetics
- CDC2 Protein Kinase/metabolism
- Cell Nucleolus/metabolism
- Cell Nucleus/metabolism
- Cytoplasm/metabolism
- Disease Models, Animal
- Dogs
- G2 Phase/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- HeLa Cells
- Humans
- Mammary Glands, Animal/enzymology
- Mammary Glands, Animal/pathology
- Mammary Neoplasms, Experimental/enzymology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Okadaic Acid/pharmacology
- Protein Transport
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- S Phase/drug effects
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Felicia Migone
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, Alabama 36849, USA
| | | | | | | |
Collapse
|
46
|
Fernando DD. Characterization of pollen tube development inPinus strobus (Eastern white pine) through proteomic analysis of differentially expressed proteins. Proteomics 2005; 5:4917-26. [PMID: 16247732 DOI: 10.1002/pmic.200500009] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The differentially expressed proteins in pollen tubes indicate their specific roles in this stage of male gametophyte development. To isolate these proteins, 2-DE was done using ungerminated pollen and 2-day-old pollen tubes of Pinus strobus. Results show that 645 and 647 protein spots were clearly resolved from pollen grains and pollen tubes, respectively. Thirty-eight protein spots were expressed only in pollen tubes, while 19 increased in intensity. MALDI-TOF MS was used to generate tryptic peptide masses that were submitted to Mascot for identification. Of the differentially expressed proteins, 12% matched with hypothetical proteins, 33% did not hit any protein, and for the 55%, a putative function was assigned based on similarity of sequences with previously characterized proteins. Therefore, pollen tube development can be characterized by the cellular activities that involve metabolism, stress/defense response, gene regulation, signal transduction, and cell wall formation. This study expands our understanding of the changes in protein expression associated with pollen tube development and provides insights into the molecular programs that separate the development of the pollen tubes from pollen grains. This is the first report that describes a global analysis of differentially expressed proteins from the pollen tube of any seed plant.
Collapse
Affiliation(s)
- Danilo D Fernando
- Department of Environmental and Forest Biology, State University of New York College of Environmental Science and Forestry, 461 Illick Hall, I Forestry Drive, Syracuse, NY 13210, USA.
| |
Collapse
|
47
|
Malumbres M, Barbacid M. Mammalian cyclin-dependent kinases. Trends Biochem Sci 2005; 30:630-41. [PMID: 16236519 DOI: 10.1016/j.tibs.2005.09.005] [Citation(s) in RCA: 890] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2005] [Revised: 08/11/2005] [Accepted: 09/15/2005] [Indexed: 02/02/2023]
Abstract
Cyclin-dependent kinases (Cdks) are the catalytic subunits of a family of mammalian heterodimeric serine/threonine kinases that have been implicated in the control of cell-cycle progression, transcription and neuronal function. Recent genetic evidence obtained with gene-targeted mice has shown that Cdk4 and Cdk6 are not needed for entry into the cell cycle after mitogenic stimuli and organogenesis; however, they are essential for the proliferation of some endocrine and hematopoietic cells. Cdk2 is also dispensable for the mitotic cell cycle. Indeed, mice without Cdk2 are normal except for their complete sterility: unexpectedly, Cdk2 is crucial for the first meiotic division of male and female germ cells. These findings have important implications both for our current understanding of the role of Cdks in regulating the mammalian cell cycle and for their potential use as therapeutic targets in cancer.
Collapse
Affiliation(s)
- Marcos Malumbres
- Molecular Oncology Programme, Centro Nacional de Investigaciones Oncológicas, Melchor Fernández Almagro 3, E-28029 Madrid, Spain.
| | | |
Collapse
|
48
|
Shieh JC, White A, Cheng YC, Rosamond J. Identification and functional characterization of Candida albicans CDC4. J Biomed Sci 2005; 12:913-24. [PMID: 16228290 DOI: 10.1007/s11373-005-9027-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2005] [Accepted: 08/17/2005] [Indexed: 10/25/2022] Open
Abstract
The CDC4 gene of Saccharomyces cerevisiae encodes an essential function that is required for G1-S and G2-M transitions during mitosis and at various stages during meiosis. We have isolated a functional homologue of CDC4 (CaCDC4) from the pathogenic yeast Candida albicans by complementing the S. cerevisiae cdc4-3 mutation with CaCDC4 expressed from its own promoter on a single-copy vector. The predicted product of CaCDC4 has 37% overall identity to the S. cerevisiae Cdc4 protein, although this identity is biased towards the C-terminal region of the two proteins which contains eight copies of the degenerate WD-40 motif, an element found in proteins that regulate diverse biological processes and an F-box domain proximal to the first iteration of the WD-40 motif. Both the F-box domain and WD-40 motifs appear necessary for the mitotic functions of Cdc4 in both yeasts. In contrast to its conserved role in mitosis, C. albicans CDC4 is unable to rescue the meiotic deficiency in a S. cerevisiae cdc4 homozygous diploid under restrictive conditions, even when expressed from an efficient S. cerevisiae promoter. In opposition to S. cerevisiae CDC4 being essential, C. albicans CDC4 appears to be nonessential and in its absence is critical for filamentous growth in C. albicans.
Collapse
Affiliation(s)
- Jia-Ching Shieh
- Department of Life Sciences, Chung Shan Medical University, No. 110, Sec. 1, Janguo N. Road, Taichung City, Taiwan, 40242, ROC.
| | | | | | | |
Collapse
|
49
|
Lyng H, Landsverk KS, Kristiansen E, DeAngelis PM, Ree AH, Myklebost O, Hovig E, Stokke T. Response of malignant B lymphocytes to ionizing radiation: gene expression and genotype. Int J Cancer 2005; 115:935-42. [PMID: 15723354 DOI: 10.1002/ijc.20962] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The human malignant B-lymphocyte cell lines Reh and U698 show arrest in G2 phase after ionizing radiation (IR), but only Reh cells arrest in G1 phase and die by apoptosis. We have used cDNA microarrays to measure changes in gene expression at 2, 4 and 6 hr after irradiation of Reh and U698 cells with 0.5 and 4 Gy in order to begin exploring the molecular mechanisms underlying the phenotypic changes. We also investigated whether gene expression changes could be caused by possible aberrations of genes, as measured by comparative genomic hybridization. Reh cells showed upregulation of CDKN1A that likely mediated the G1 arrest. In contrast, U698 cells have impaired function of TP53 protein and no activation of CDKN1A, suppressing the arrest in G1. The G2 arrest in both cell lines was likely due to repression of PLK1 and/or CCNF. IR-induced apoptosis in Reh cells was probably mediated by TP53 and CDKN1A, whereas a high expression level of MCL1, caused by gene amplification, and activation of the NFKB pathway may have suppressed the apoptotic response in U698 cells. Genes suggested to be involved in apoptosis were activated long before this phenotype was detectable and showed the same temporal expression profiles as genes involved in cell cycle arrest. Our results suggest that differences in functionality and/or copy number of several genes involved in IR-regulated pathways contributed to the phenotypic differences between Reh and U698 cells after IR, and that multiple molecular factors control the radiation response of malignant B lymphocytes.
Collapse
Affiliation(s)
- Heidi Lyng
- Department of Radiation Biology, The Norwegian Radium Hospital, Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Hogarth C, Itman C, Jans DA, Loveland KL. Regulated nucleocytoplasmic transport in spermatogenesis: a driver of cellular differentiation? Bioessays 2005; 27:1011-25. [PMID: 16163727 DOI: 10.1002/bies.20289] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This review explores the hypothesis that regulation of nucleocytoplasmic shuttling is a means of driving differentiation, using spermatogenesis as a model. The transition from undifferentiated spermatogonial stem cell to terminally differentiated spermatozoon is, at its most basic, a change in the repertoire of expressed genes. To effect this, the complement of nuclear proteins, such as transcription factors and chromatin remodelling components must change. Current knowledge of the nuclear proteins and nucleocytoplasmic transport machinery relevant to spermatogenesis is consolidated in this review, and their functional linkages are highlighted not only as a means of regulating nuclear protein composition, but also as a key mechanism regulating gene transcription and hence cell fate. Through this, we hypothesize that male germ cell differentiation is mediated through regulation of nuclear transport machinery components, and thereby of the access of critical factors to the nucleus. The importance of nucleocytoplasmic trafficking to male germ cell differentiation is discussed, using the sex-determining factors Sry and SOX9, cell cycle regulators, CREM and cofactors and the Smads as specific examples, together with the roles in gametogenesis for particular nuclear transport factors in Caenorhabditis elegans and Drosophila.
Collapse
Affiliation(s)
- Cathryn Hogarth
- Monash Institute of Medical Research, Monash University, Melbourne, Australia
| | | | | | | |
Collapse
|