1
|
Moore XTR, Gheghiani L, Fu Z. The Role of Polo-Like Kinase 1 in Regulating the Forkhead Box Family Transcription Factors. Cells 2023; 12:cells12091344. [PMID: 37174744 PMCID: PMC10177174 DOI: 10.3390/cells12091344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine kinase with more than 600 phosphorylation substrates through which it regulates many biological processes, including mitosis, apoptosis, metabolism, RNA processing, vesicle transport, and G2 DNA-damage checkpoint recovery, among others. Among the many PLK1 targets are members of the FOX family of transcription factors (FOX TFs), including FOXM1, FOXO1, FOXO3, and FOXK1. FOXM1 and FOXK1 have critical oncogenic roles in cancer through their antagonism of apoptotic signals and their promotion of cell proliferation, metastasis, angiogenesis, and therapeutic resistance. In contrast, FOXO1 and FOXO3 have been identified to have broad functions in maintaining cellular homeostasis. In this review, we discuss PLK1-mediated regulation of FOX TFs, highlighting the effects of PLK1 on the activity and stability of these proteins. In addition, we review the prognostic and clinical significance of these proteins in human cancers and, more importantly, the different approaches that have been used to disrupt PLK1 and FOX TF-mediated signaling networks. Furthermore, we discuss the therapeutic potential of targeting PLK1-regulated FOX TFs in human cancers.
Collapse
Affiliation(s)
- Xavier T R Moore
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Lilia Gheghiani
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Zheng Fu
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA 23298, USA
| |
Collapse
|
2
|
Iliaki S, Beyaert R, Afonina IS. Polo-like kinase 1 (PLK1) signaling in cancer and beyond. Biochem Pharmacol 2021; 193:114747. [PMID: 34454931 DOI: 10.1016/j.bcp.2021.114747] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023]
Abstract
PLK1 is an evolutionary conserved Ser/Thr kinase that is best known for its role in cell cycle regulation and is expressed predominantly during the G2/S and M phase of the cell cycle. PLK1-mediated phosphorylation of specific substrates controls cell entry into mitosis, centrosome maturation, spindle assembly, sister chromatid cohesion and cytokinesis. In addition, a growing body of evidence describes additional roles of PLK1 beyond the cell cycle, more specifically in the DNA damage response, autophagy, apoptosis and cytokine signaling. PLK1 has an indisputable role in cancer as it controls several key transcription factors and promotes cell proliferation, transformation and epithelial-to-mesenchymal transition. Furthermore, deregulation of PLK1 results in chromosome instability and aneuploidy. PLK1 is overexpressed in many cancers, which is associated with poor prognosis, making PLK1 an attractive target for cancer treatment. Additionally, PLK1 is involved in immune and neurological disorders including Graft versus Host Disease, Huntington's disease and Alzheimer's disease. Unfortunately, newly developed small compound PLK1 inhibitors have only had limited success so far, due to low therapeutic response rates and toxicity. In this review we will highlight the current knowledge about the established roles of PLK1 in mitosis regulation and beyond. In addition, we will discuss its tumor promoting but also tumor suppressing capacities, as well as the available PLK1 inhibitors, elaborating on their efficacy and limitations.
Collapse
Affiliation(s)
- Styliani Iliaki
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Rudi Beyaert
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.
| | - Inna S Afonina
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| |
Collapse
|
3
|
Chow AKM, Yau SWL, Ng L. Novel molecular targets in hepatocellular carcinoma. World J Clin Oncol 2020; 11:589-605. [PMID: 32879846 PMCID: PMC7443834 DOI: 10.5306/wjco.v11.i8.589] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/04/2020] [Accepted: 06/20/2020] [Indexed: 02/06/2023] Open
Abstract
Globally, hepatocellular carcinoma (HCC) is a leading cause of cancer and cancer-related deaths. The therapeutic efficacy of locoregional and systemic treatment in patients with advanced HCC remains low, which results in a poor prognosis. The development of sorafenib for the treatment of HCC has resulted in a new era of molecular targeted therapy for this disease. However, the median overall survival was reported to be barely higher in the sorafenib treatment group than in the control group. Hence, in this review we describe the importance of developing more effective targeted therapies for the management of advanced HCC. Recent investigations of molecular signaling pathways in several cancers have provided some insights into developing molecular therapies that target critical members of these signaling pathways. Proteins involved in the Hedgehog and Notch signaling pathways, Polo-like kinase 1, arginine, histone deacetylases and Glypican-3 can be potential targets in the treatment of HCC. Monotherapy has limited therapeutic efficacy due to the development of inhibitory feedback mechanisms and induction of chemoresistance. Thus, emphasis is now on the development of personalized and combination molecular targeted therapies that can serve as ideal therapeutic strategies for improved management of HCC.
Collapse
Affiliation(s)
- Ariel Ka-Man Chow
- School of Nursing and Health Studies, The Open University of Hong Kong, Hong Kong, China
| | - Simon Wing-Lung Yau
- School of Nursing and Health Studies, The Open University of Hong Kong, Hong Kong, China
| | - Lui Ng
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
4
|
Zhao D, Shun E, Ling F, Liu Q, Warsi A, Wang B, Zhou Q, Zhu C, Zheng H, Liu K, Zheng X. Plk2 Regulated by miR-128 Induces Ischemia-Reperfusion Injury in Cardiac Cells. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 19:458-467. [PMID: 31902745 PMCID: PMC6948232 DOI: 10.1016/j.omtn.2019.11.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/11/2019] [Accepted: 11/24/2019] [Indexed: 12/19/2022]
Abstract
Ischemia-reperfusion (I/R) injury occurs during cardiac surgery and is the major factor leading to heart dysfunction and heart failure. Our previous study showed that gene and microRNA expression profiles are altered in heart grafts with extended I/R injury. In this study, we, for the first time, demonstrated that I/R injury upregulates the expression of Polo-like kinase 2 (Plk2) but decreases miR-128 expression in heart cells both in vitro and in vivo. Silencing Plk2 using small interfering RNA (siRNA) protects cells from Antimycin A-induced cell apoptosis/death. Silencing Plk2 also decreases phosphorylated p65 expression but increases Angiopoietin 1 expression. In addition, Plk2 is negatively regulated by miR-128. miR-128 exerts a protective effect on cell apoptosis similar to Plk2 siRNA in response to I/R stress. Methylation inhibitor 5-azacytidine (5-AZ) increases the expression of miR-128 and subsequently reduces Plk2 expression and cell apoptosis. In conclusion, this study demonstrated that Plk2 regulated by miR-128 induces cell apoptosis/death in response to I/R stress through activation of the nuclear factor κB (NF-κB) signal pathway. miR-128 and Plk2 are new targets for preventing cardiac I/R injury or oxidative stress-mediated injury.
Collapse
Affiliation(s)
- Duo Zhao
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China; Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada; Department of Cardiovascular Surgery, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Edward Shun
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Fengjun Ling
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Qing Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Ayesha Warsi
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Bowen Wang
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China; Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Qinfeng Zhou
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Cuilin Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China; Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Hao Zheng
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China.
| | - Xiufen Zheng
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada; Department of Surgery, Western University, London, ON, Canada; Department of Oncology, Western University, London, ON, Canada; Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
5
|
Branter J, Basu S, Smith S. Tumour treating fields in a combinational therapeutic approach. Oncotarget 2018; 9:36631-36644. [PMID: 30564303 PMCID: PMC6290966 DOI: 10.18632/oncotarget.26344] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 10/24/2018] [Indexed: 12/15/2022] Open
Abstract
The standard of care for patients with newly diagnosed Glioblastoma multiforme (GBM) has remained unchanged since 2005, with patients undergoing maximal surgical resection, followed by radiotherapy plus concomitant and maintenance Temozolomide. More recently, Tumour treating fields (TTFields) therapy has become FDA approved for adult recurrent and adult newly-diagnosed GBM following the EF-11 and EF-14 trials, respectively. TTFields is a non-invasive anticancer treatment which utilizes medium frequency alternating electric fields to target actively dividing cancerous cells. TTFields selectively targets cells within mitosis through interacting with key mitotic proteins to cause mitotic arrest and cell death. TTFields therapy presents itself as a candidate for the combinational therapy route due to the lack of overlapping toxicities associated with electric fields. Here we review current literature pertaining to TTFields in combination with alkylating agents, radiation, anti-angiogenics, mitotic inhibitors, immunotherapies, and also with novel agents. This review highlights the observed synergistic and additive effects of combining TTFields with various other therapies, as well highlighting the strategies relating to combinations with electric fields.
Collapse
Affiliation(s)
- Joshua Branter
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| | - Surajit Basu
- Queen's Medical Centre, Department of Neurosurgery, Nottingham, UK
| | - Stuart Smith
- Children's Brain Tumour Research Centre, School of Medicine, University of Nottingham, Queen's Medical Centre, Nottingham, UK
| |
Collapse
|
6
|
Bowman BM, Sebolt KA, Hoff BA, Boes JL, Daniels DL, Heist KA, Galbán CJ, Patel RM, Zhang J, Beer DG, Ross BD, Rehemtulla A, Galbán S. Phosphorylation of FADD by the kinase CK1α promotes KRASG12D-induced lung cancer. Sci Signal 2015; 8:ra9. [PMID: 25628462 PMCID: PMC4416214 DOI: 10.1126/scisignal.2005607] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Genomic amplification of the gene encoding and phosphorylation of the protein FADD (Fas-associated death domain) is associated with poor clinical outcome in lung cancer and in head and neck cancer. Activating mutations in the guanosine triphosphatase RAS promotes cell proliferation in various cancers. Increased abundance of phosphorylated FADD in patient-derived tumor samples predicts poor clinical outcome. Using immunohistochemistry analysis and in vivo imaging of conditional mouse models of KRAS(G12D)-driven lung cancer, we found that the deletion of the gene encoding FADD suppressed tumor growth, reduced the proliferative index of cells, and decreased the activation of downstream effectors of the RAS-MAPK (mitogen-activated protein kinase) pathway that promote the cell cycle, including retinoblastoma (RB) and cyclin D1. In mouse embryonic fibroblasts, the induction of mitosis upon activation of KRAS required FADD and the phosphorylation of FADD by CK1α (casein kinase 1α). Deleting the gene encoding CK1α in KRAS mutant mice abrogated the phosphorylation of FADD and suppressed lung cancer development. Phosphorylated FADD was most abundant during the G2/M phase of the cell cycle, and mass spectrometry revealed that phosphorylated FADD interacted with kinases that mediate the G2/M transition, including PLK1 (Polo-like kinase 1), AURKA (Aurora kinase A), and BUB1 (budding uninhibited by benzimidazoles 1). This interaction was decreased in cells treated with a CKI-7, a CK1α inhibitor. Therefore, as the kinase that phosphorylates FADD downstream of RAS, CK1α may be a therapeutic target for KRAS-driven lung cancer.
Collapse
Affiliation(s)
- Brittany M Bowman
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Katrina A Sebolt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Benjamin A Hoff
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jennifer L Boes
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Kevin A Heist
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Craig J Galbán
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rajiv M Patel
- Departments of Pathology and Dermatology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianke Zhang
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - David G Beer
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brian D Ross
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA. Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alnawaz Rehemtulla
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Stefanie Galbán
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Barton O, Naumann SC, Diemer-Biehs R, Künzel J, Steinlage M, Conrad S, Makharashvili N, Wang J, Feng L, Lopez BS, Paull TT, Chen J, Jeggo PA, Löbrich M. Polo-like kinase 3 regulates CtIP during DNA double-strand break repair in G1. ACTA ACUST UNITED AC 2014; 206:877-94. [PMID: 25267294 PMCID: PMC4178966 DOI: 10.1083/jcb.201401146] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Plk3 phosphorylates CtIP in G1 in a damage-inducible manner and is required with CtIP for the repair of complex double-strand breaks and regulation of resection-mediated end-joining pathways. DNA double-strand breaks (DSBs) are repaired by nonhomologous end joining (NHEJ) or homologous recombination (HR). The C terminal binding protein–interacting protein (CtIP) is phosphorylated in G2 by cyclin-dependent kinases to initiate resection and promote HR. CtIP also exerts functions during NHEJ, although the mechanism phosphorylating CtIP in G1 is unknown. In this paper, we identify Plk3 (Polo-like kinase 3) as a novel DSB response factor that phosphorylates CtIP in G1 in a damage-inducible manner and impacts on various cellular processes in G1. First, Plk3 and CtIP enhance the formation of ionizing radiation-induced translocations; second, they promote large-scale genomic deletions from restriction enzyme-induced DSBs; third, they are required for resection and repair of complex DSBs; and finally, they regulate alternative NHEJ processes in Ku−/− mutants. We show that mutating CtIP at S327 or T847 to nonphosphorylatable alanine phenocopies Plk3 or CtIP loss. Plk3 binds to CtIP phosphorylated at S327 via its Polo box domains, which is necessary for robust damage-induced CtIP phosphorylation at S327 and subsequent CtIP phosphorylation at T847.
Collapse
Affiliation(s)
- Olivia Barton
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Steffen C Naumann
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Ronja Diemer-Biehs
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Julia Künzel
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Monika Steinlage
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Sandro Conrad
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| | - Nodar Makharashvili
- The Howard Hughes Medical Institute, Department of Molecular Genetics and Microbiology, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712
| | - Jiadong Wang
- Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Lin Feng
- Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Bernard S Lopez
- Centre National de la Recherche Scientifique Unité Mixte de Recherche 8200, Institut de Cancérologie Gustave-Roussy, Université Paris-Sud, F-94805 Villejuif, France
| | - Tanya T Paull
- The Howard Hughes Medical Institute, Department of Molecular Genetics and Microbiology, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX 78712
| | - Junjie Chen
- Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Penny A Jeggo
- Genome Damage and Stability Centre, University of Sussex, Brighton BN1 9RQ, England, UK
| | - Markus Löbrich
- Radiation Biology and DNA Repair, Darmstadt University of Technology, 64287 Darmstadt, Germany
| |
Collapse
|
8
|
Chastain PD, Brylawski BP, Zhou YC, Rao S, Chu H, Ibrahim JG, Kaufmann WK, Cordeiro-Stone M. DNA damage checkpoint responses in the S phase of synchronized diploid human fibroblasts. Photochem Photobiol 2014; 91:109-16. [PMID: 25316620 PMCID: PMC4303954 DOI: 10.1111/php.12361] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 10/08/2014] [Indexed: 01/05/2023]
Abstract
We investigated the hypothesis that the strength of the activation of the intra-S DNA damage checkpoint varies within the S phase. Synchronized diploid human fibroblasts were exposed to either 0 or 2.5 J m−2 UVC in early, mid- and late-S phase. The endpoints measured were the following: (1) radio-resistant DNA synthesis (RDS), (2) induction of Chk1 phosphorylation, (3) initiation of new replicons and (4) length of replication tracks synthesized after irradiation. RDS analysis showed that global DNA synthesis was inhibited by approximately the same extent (30 ± 12%), regardless of when during S phase the fibroblasts were exposed to UVC. Western blot analysis revealed that the UVC-induced phosphorylation of checkpoint kinase 1 (Chk1) on serine 345 was high in early and mid S but 10-fold lower in late S. DNA fiber immunostaining studies indicated that the replication fork displacement rate decreased in irradiated cells at the three time points examined; however, replicon initiation was inhibited strongly in early and mid S, but this response was attenuated in late S. These results suggest that the intra-S checkpoint activated by UVC-induced DNA damage is not as robust toward the end of S phase in its inhibition of the latest firing origins in human fibroblasts.
Collapse
Affiliation(s)
- Paul D Chastain
- College of Osteopathic Medicine, William Carey University, Hattiesburg, MS
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Zhao XY, Nie CL, Liang SF, Yuan Z, Deng HX, Wei YQ. Enhanced gemcitabine-mediated cell killing of human lung adenocarcinoma by vector-based RNA interference against PLK1. Biomed Pharmacother 2012; 66:597-602. [PMID: 23153503 DOI: 10.1016/j.biopha.2012.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 01/04/2012] [Indexed: 02/05/2023] Open
Abstract
Specific PLK1 silencing may be an effective gene therapy modality of treating PLK1-overexpressed cancers. In this study, we first explored the anticancer efficacy of three different short hairpin-expressing plasmids targeting PLK1 in animal model, and then determined the combination therapy effect of gemcitabine with PLK1-shRNA as an adjuvant. Transfection of the PLK1-shRNAs to A549 lung cancer cells induced significant PLK1 depletion, growth inhibition and apoptosis. In vivo administration of PLK1-shRNA constructs to tumor-bearing mice resulted in xenograft regression. Moreover, the combination of PLK1-shRNA plus low-dose gemcitabine (GEM) produced an additive antitumor activity on the lung tumors owing to an inhibition of cancer cell survival and augmented apoptosis. These results indicated a feasible bio-chemotherapeutic strategy for cancer.
Collapse
Affiliation(s)
- Xin-Yu Zhao
- State Key Laboratory of Biotherapy and Cancer Centre, West China Hospital, Sichuan University, Chengdu, China
| | | | | | | | | | | |
Collapse
|
10
|
Han DP, Zhu QL, Cui JT, Wang PX, Qu S, Cao QF, Zong YP, Feng B, Zheng MH, Lu AG. Polo-like kinase 1 is overexpressed in colorectal cancer and participates in the migration and invasion of colorectal cancer cells. Med Sci Monit 2012; 18:BR237-46. [PMID: 22648245 PMCID: PMC3560731 DOI: 10.12659/msm.882900] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 11/13/2011] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Polo-like kinase 1 (PLK1) is an important molecule in proliferation of many human cancers. The aim of study is to clarify the expression patterns and potential function of PLK1 in colorectal cancers. MATERIAL/METHODS Fifty-six colorectal cancers samples were collected and arranged onto a tissue array and the expression of PLK1 were detected by immunohistochemistry and correlated with clinico-pathological characteristics and expression of PCNA. Expression of PLK1 in 9 colorectal cancer cells lines was investigated by RT-PCR and Western blot, then SW1116 cells lines were treated with PLK1 siRNA and the efficiency was examined by Western blot. Transwell test was applied to detect the migration and invasion capability of cancer cells by counting the number of cells passing through the membranes. Cell proliferation and apoptosis were examined by Cell Counting Kit-8 (CCK-8) and Annexin-V Kit. RESULTS PLK1 was positively expressed in 73.2% (41/56) of colorectal cancers tissues, but in only 3.6% (2/56) of normal tissues, and was associated with Duke's stage (P<0.01), tumor size (P<0.01), invasion extent (P<0.05) and lymphatic metastasis (P<0.01). The expression of PLK1 was correlated with expression of PCNA (R=0.553, P<0.01). PLK1 was inhibited in SW1116 cells by treating with PLK1 siRNA oligos, which resulted in a decreased number of cells passing through the membrane as compared with control groups (P<0.01) at 24 hours after transfection. Cell proliferation was inhibited from 48 hours after transfection, while cells apoptosis was induced from 72 hours after transfection. CONCLUSIONS PLK1 could be a progression marker for colorectal cancer patients and PLK1 depletion can inhibit migration and invasion capability of colorectal cancer cells SW1116, suggesting that PLK1 might be involved in metastasis and invasion of colorectal cancer. Therapeutic strategies targeting PLK1 may be a new approach to colorectal cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ai-guo Lu
- Lu Ai-Guo, Department of General Surgery, Shanghai Ruijin Hospital, Shanghai Minimally Invasive Surgery Center, 197 Ruijin Er Rd, Shanghai 200025, China, e-mail:
| |
Collapse
|
11
|
Yoon HE, Kim SA, Choi HS, Ahn MY, Yoon JH, Ahn SG. Inhibition of Plk1 and Pin1 by 5'-nitro-indirubinoxime suppresses human lung cancer cells. Cancer Lett 2012; 316:97-104. [PMID: 22115795 DOI: 10.1016/j.canlet.2011.10.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 10/15/2011] [Accepted: 10/20/2011] [Indexed: 10/15/2022]
Abstract
A novel indirubin derivative, 5'-nitro-indirubinoxime (5'-NIO), exhibits a strong anti-cancer activity against human cancer cells. Here, the 5'-NIO-mediated G1 cell cycle arrest in lung cancer cells was associated with a decrease in protein levels of polo-like kinase 1 (Plk1) and peptidyl-prolyl cis/trans isomerase Pin1. Treatment with Plk1 siRNA or Pin1 inhibitor effectively inhibited the Rb phosphorylation, suggesting their regulatory role at G1 phase. In addition, the overexpression of Plk1 or Pin1 inhibited apoptotic signals following the cleavage of PARP in 5'-NIO-treated cells. These findings suggest that 5'-NIO have potential anti-cancer efficacy through the inhibition of Plk1 or/and Pin1 expression.
Collapse
Affiliation(s)
- Hyo-Eun Yoon
- Department of Pathology, Chosun University College of Dentistry, Gwangju 501-759, Republic of Korea
| | | | | | | | | | | |
Collapse
|
12
|
Lin YC, Sun SH, Wang FF. Suppression of Polo like kinase 1 (PLK1) by p21(Waf1) mediates the p53-dependent prevention of caspase-independent mitotic death. Cell Signal 2011; 23:1816-23. [PMID: 21726628 DOI: 10.1016/j.cellsig.2011.06.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 06/20/2011] [Indexed: 01/10/2023]
Abstract
Polo-like kinase 1 (Plk1) plays key roles in many aspects of mitosis. We have previously shown that induction of p21(Waf1) by p53 is responsible for protection of cells against adriamycin-induced polyploidy formation and mitotic catastrophe. Here we show that adriamycin treatment suppressed Plk1 expression in a p53- and p21(Waf1)-dependent manner. Ablation of p21(Waf1) inhibited the adriamycin-induced p53 activation, and this inhibition was alleviated by knockdown of Plk1, suggesting that p21(Waf1)-dependent suppression of Plk1 expression is responsible for maintaining p53 activation during stress response. Plk1 associated with p53 and disrupted its interaction with target gene promoters in cells treated with adriamycin. Overexpression of Plk1 inhibited the p53-mediated prevention of caspase-independent mitotic death, but not polyploidy formation, in adriamycin-treated cells. Together our results indicate that suppression of Plk1 by p21(Waf1) is responsible for p53-dependent protection against adriamycin-induced caspase-independent mitotic death.
Collapse
Affiliation(s)
- Yi-Cheng Lin
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, 11221, Taiwan
| | | | | |
Collapse
|
13
|
Strebhardt K. Multifaceted polo-like kinases: drug targets and antitargets for cancer therapy. Nat Rev Drug Discov 2010; 9:643-60. [PMID: 20671765 DOI: 10.1038/nrd3184] [Citation(s) in RCA: 547] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The polo-like kinase 1 (PLK1) acts in concert with cyclin-dependent kinase 1-cyclin B1 and Aurora kinases to orchestrate a wide range of critical cell cycle events. Because PLK1 has been preclinically validated as a cancer target, small-molecule inhibitors of PLK1 have become attractive candidates for anticancer drug development. Although the roles of the closely related PLK2, PLK3 and PLK4 in cancer are less well understood, there is evidence showing that PLK2 and PLK3 act as tumour suppressors through their functions in the p53 signalling network, which guards the cell against various stress signals. In this article, recent insights into the biology of PLKs will be reviewed, with an emphasis on their role in malignant transformation, and progress in the development of small-molecule PLK1 inhibitors will be examined.
Collapse
Affiliation(s)
- Klaus Strebhardt
- Department of Obstetrics and Gynaecology, School of Medicine, J.W. Goethe University, Theodor Stern Kai 7, 60590 Frankfurt, Germany.
| |
Collapse
|
14
|
ITO J, YOSHIDA T, KASAI Y, WAKAI T, PARYS JB, FISSORE RA, KASHIWAZAKI N. Phosphorylation of inositol 1,4,5-triphosphate receptor 1 duringin vitromaturation of porcine oocytes. Anim Sci J 2010; 81:34-41. [DOI: 10.1111/j.1740-0929.2009.00699.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
15
|
Chopra P, Sethi G, Dastidar SG, Ray A. Polo-like kinase inhibitors: an emerging opportunity for cancer therapeutics. Expert Opin Investig Drugs 2010; 19:27-43. [PMID: 20001553 DOI: 10.1517/13543780903483191] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD The Polo-like kinase (Plk) family has emerged as an important regulator in cell cycle progression. Plks belong to a family of serine/threonine kinases and exist in four isoforms Plk1- 4. However, only one of these isoforms, Plk1, is shown to be involved in the activation of Cdc2, chromosome segregation, centrosome maturation, bipolar spindle formation and execution of cytokinesis. The activity of Plk1 is elevated in tissues and cells with a high mitotic index. In patients, Plk1 is overexpressed in tumors including those derived from lung, breast, colon, pancreas, prostate and ovary. Plk1 depletion is associated with the decrease in cell viability and induction of apoptosis in various cancerous cells. Several Plk1 inhibitors are in different phases of clinical development for anticancer therapy. AREAS COVERED IN THIS REVIEW The focus of present review is to highlight Plk1 as a promising therapeutic approach for the treatment of cancer. The review discusses the role of Plk1 in cancer and the current status of Plk1 inhibitors, as well as highlighting the possible beneficial effect of inhibition of Plk1 as compared to other mitotic targets. WHAT THE READER WILL GAIN Readers will get a comprehensive overview of Plk1 as a novel anticancer drug target. This review will also update readers about the progress made in the field of Plk1 inhibitors. TAKE HOME MESSAGE The current literature about Plk1 inhibitors and knockout studies favor Plk1 inhibition as a potential antitumor therapy.
Collapse
Affiliation(s)
- Puneet Chopra
- New Drug Discovery Research, Department of Pharmacology, Ranbaxy Research Laboratories, Gurgaon-122001-Haryana, India.
| | | | | | | |
Collapse
|
16
|
Imai H, Sugimoto K, Isobe Y, Sasaki M, Yasuda H, Takeuchi K, Nakamura S, Kojima Y, Tomomatsu J, Oshimi K. Absence of tumor-specific over-expression of Polo-like kinase 1 (Plk1) in major non-Hodgkin lymphoma and relatively low expression of Plk1 in nasal NK/T cell lymphoma. Int J Hematol 2009; 89:673-8. [PMID: 19452252 DOI: 10.1007/s12185-009-0325-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 04/13/2009] [Accepted: 04/15/2009] [Indexed: 12/15/2022]
Abstract
Based on the presence of the tumor-specific over-expression of Plk1 (polo-like kinases) in various malignancies, we examined Plk1 expression in nine cases of reactive follicular hyperplasia (RFH), 42 of diffuse large B cell lymphoma (DLBCL), 16 of follicular lymphoma (FL), and 10 of nasal NK/T lymphoma. There was no significant difference in the Plk1-positive cell percentage between RFH and DLBCL. The Plk1-positive cell percentage ranged from 6 to 20% with a median of 12.9% in DLBCL. In FL, Plk1-positivity was at most 7%. Plk1-positivity in nasal NK/T cell lymphoma (4.7-14.1% with a median of 9.2%) was significantly higher than that of FL and tended to be lower than DLBCL (p < 0.001, p = 0.05, respectively). Although a strong correlation between positive cell percentages for Plk1 and Ki-67 in these three lymphomas specified Plk1 as a proliferation marker (r = 0.83-0.91), the Plk1-positive cell percentage relative to the other proliferation markers tended to be particularly low in nasal NK/T cell lymphoma. In 41 cases of DLBCL, the positive cell percentages of Plk1 and Ki-67 were both correlated with overall survival. The 4-year overall survival rates by Kaplan-Meier analysis for Plk1-negative and positive patients were 80 and 38%, respectively (p = 0.02).
Collapse
MESH Headings
- Biomarkers
- Cell Cycle Proteins/analysis
- Cell Cycle Proteins/genetics
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Humans
- Ki-67 Antigen/analysis
- Lymphoma, Extranodal NK-T-Cell/chemistry
- Lymphoma, Extranodal NK-T-Cell/mortality
- Lymphoma, Follicular/chemistry
- Lymphoma, Large B-Cell, Diffuse/chemistry
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Non-Hodgkin/chemistry
- Lymphoma, Non-Hodgkin/mortality
- Neoplasm Proteins/analysis
- Neoplasm Proteins/genetics
- Nose Neoplasms/chemistry
- Nose Neoplasms/mortality
- Prognosis
- Protein Serine-Threonine Kinases/analysis
- Protein Serine-Threonine Kinases/genetics
- Proto-Oncogene Proteins/analysis
- Proto-Oncogene Proteins/genetics
- Survival Analysis
- Polo-Like Kinase 1
Collapse
Affiliation(s)
- Hidenori Imai
- Department of Internal Medicine, Division of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Nie JY, Huang JA, Tang GD, Liu SQ. Effect of sulindac on apoptosis and related gene expression profile of human colon cancer cells. Shijie Huaren Xiaohua Zazhi 2008; 16:2826-2830. [DOI: 10.11569/wcjd.v16.i25.2826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the changes of cell apoptosis and gene expression profile of human colon carcinoma cell line Lovo after sulindac treatment.
METHODS: Transmission electron microscopy and flow cytometry were used to observe the apoptosis changes of LoVo cells 48 and 72 h after sulindac treatment; meanwhile, cDNA microarray was used to detect the genes differetially expressed in LoVo cells.
RESULTS: Apoptotic bodies were found and the apoptotic rates of LoVo cells increased greatly after treatment with 0.6, 0.9 and 1.2 mmol/L sulindac in comparison with those of control cells (48 h: 4.2 ± 1.04, 4.26 ± 0.28, 7.51 ± 2.09 vs 1.81 ± 0.91; 72 h: 6.21 ± 0.56, 7.48 ± 1.45, 10.40 ± 1.30 vs 2.06 ± 1.43; all P < 0.05). Hybridization with cDNA microarray containing 17101 genes screened 1013 differetially expressed genes, of which 178 genes (17.87%) were associated with cell apoptosis. Of the 178 genes, 82 were up-regulated while 96 were down-regulated.
CONCLUSION: Sulindac can induce apoptosis of LoVo cells, and its mechanism may attribute to up-regulation or down-regulation of some apoptosis-related genes.
Collapse
|
18
|
Ismail IA, Kang KS, Lee HA, Kim JW, Sohn YK. Genistein-induced neuronal apoptosis and G2/M cell cycle arrest is associated with MDC1 up-regulation and PLK1 down-regulation. Eur J Pharmacol 2007; 575:12-20. [PMID: 17706963 DOI: 10.1016/j.ejphar.2007.07.039] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2007] [Revised: 07/19/2007] [Accepted: 07/23/2007] [Indexed: 01/14/2023]
Abstract
The aim of the present study is to investigate the effect of genistein on human neuroblastoma SK-N-MC cells. MTT proliferation assay, LDH cytotoxicity assay, flow cytometric analysis, real-time quantitative RT-PCR and western blotting were used to investigate the effect of genistein on cell survival, cellular toxicity, cell cycle progression, and mRNA and protein alterations of selected DNA damage-, cell cycle- and apoptosis-related genes in SK-N-MC cells. Genistein suppressed cell proliferation, increased LDH release and modulated cell cycle distribution through accumulation of cells at G2/M- and S-phase and sub-G0 (cell death) with a concurrent decrease of cells at G0/G1 phase. Genistein increased the MDC1 (Mediator of DNA damage Checkpoint protein 1), p53, p21(waf1/cip1), Cdc2 and Bax mRNA levels in a dose-dependent manner. However, PLK1 (Polo-Like Kinase 1) and Cyclin B1 mRNAs were down-regulated after genistein treatment. Furthermore, Genistein did not alter Chk2 (Checkpoint Kinase 2), Bcl-2 and Cdc25C mRNA levels. On western blotting analyses; genistein increased the protein level of MDC1, p53, p21(waf1/cip1), and Bax in a dose-dependent manner. Genistein also increased the phosphorylation of Chk2 and Cdc25C at Thr-68 and Ser-216, respectively. In addition, consistently with PLK1 down-regulation, the phosphorylation of Cdc25C at Ser-198 was markedly decreased after genistein treatment. Additionally, Chk2, Cdc25C, Cyclin B1, p-Cyclin B1 (Ser-147), and Cdc2 as well as Bcl-2 proteins were down-regulated after genistein treatment. Altogether, these results suggest for the first time the involvement of MDC1 up-regulation after genistein treatment in DNA damage-induced Chk2 activation- and PLK1 down-regulation-mediated apoptosis and cell cycle checkpoint pathways.
Collapse
Affiliation(s)
- Ismail Ahmed Ismail
- Department of Pathology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | | | | | | | | |
Collapse
|
19
|
Abstract
The polo-like kinase, Plk1, which is expressed and active in mitosis, is involved in regulation of mitotic entry, spindle pole assembly, mitotic exit, and cytokinesis [Donaldson MM, Tavares AA, Hagan IM, Nigg EA, Glover DM (2001) J Cell Sci 114:2357-2358]. In mammals, there are two other members of the polo-like kinase family that are less well understood, Plk2 and Plk3. Plk3 first was identified and cloned as an immediate early gene. Here, we report Plk3 localizes to the nucleolus and is involved in regulation of the G(1)/S phase transition. We demonstrate that the level of Plk3 protein is cell cycle regulated, peaking in G(1). We have delivered Plk3-interfering RNA with lentivirus to serum-starved cells and found that, upon serum stimulation, Plk3 is required for cyclin E expression and entry into S phase. Plk3-interfering RNA-induced Plk3 depletion resulted in a large fraction of asynchronously proliferating cells to become quiescent. We propose the Plk3 requirement in the cell cycle is fulfilled in G(1), and that once cells pass this point, they are able to complete cell division, whereas in the absence of Plk3, they fail to reenter the cell cycle. Additional data suggest that Plk3 may regulate entry into S phase in part through interaction with the phosphatase Cdc25A, because its depletion also resulted in attenuation of cyclin E expression.
Collapse
Affiliation(s)
- Wendy C. Zimmerman
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
| | - Raymond L. Erikson
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|
20
|
Winkles JA, Alberts GF. Differential regulation of polo-like kinase 1, 2, 3, and 4 gene expression in mammalian cells and tissues. Oncogene 2005; 24:260-6. [PMID: 15640841 DOI: 10.1038/sj.onc.1208219] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The four mammalian polo-like kinase (Plk) family members are critical regulators of cell cycle progression, mitosis, cytokinesis, and the DNA damage response. Research conducted to date has primarily investigated the expression patterns, structural features, substrates, and subcellular distribution of these important serine-threonine kinases. Here, we review the published data describing the regulation of Plk1, 2, 3, or 4 gene expression either during mammalian cell cycle progression or in tissue samples. These studies have demonstrated that the Plk family genes are differentially expressed following growth factor stimulation of quiescent fibroblasts. Furthermore, although Plk1 and Plk2 mRNA and protein levels are coordinately regulated during cell cycle progression, this is not the case for Plk3. In addition, the Plk1, 2 and 4 proteins have relatively short intracellular half-lives, but Plk3 is very stable. The Plk family genes are also differentially regulated in stressed cells; for example, when DNA-damaging agents are added to cycling cells, Plk1 expression decreases, but Plk2 and Plk3 expression increases. Finally, Plk1, 2, 3, and 4 are expressed to varying degrees in different human tissue types and it has been reported that Plk1 expression is increased and Plk3 expression is decreased in tumor specimens. These results indicate that the differential regulation of Plk family member gene expression is one cellular strategy for controlling Plk activity in mammalian cells.
Collapse
Affiliation(s)
- Jeffrey A Winkles
- Department of Surgery, University of Maryland Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | |
Collapse
|
21
|
Kues WA, Carnwath JW, Niemann H. From fibroblasts and stem cells: implications for cell therapies and somatic cloning. Reprod Fertil Dev 2005; 17:125-34. [PMID: 15745637 DOI: 10.1071/rd04118] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2004] [Accepted: 10/01/2004] [Indexed: 12/31/2022] Open
Abstract
Pluripotent embryonic stem cells (ESCs) from the inner cell mass of early murine and human embryos exhibit extensive self-renewal in culture and maintain their ability to differentiate into all cell lineages. These features make ESCs a suitable candidate for cell-replacement therapy. However, the use of early embryos has provoked considerable public debate based on ethical considerations. From this standpoint, stem cells derived from adult tissues are a more easily accepted alternative. Recent results suggest that adult stem cells have a broader range of potency than imagined initially. Although some claims have been called into question by the discovery that fusion between the stem cells and differentiated cells can occur spontaneously, in other cases somatic stem cells have been induced to commit to various lineages by the extra- or intracellular environment. Recent data from our laboratory suggest that changes in culture conditions can expand a subpopulation of cells with a pluripotent phenotype from primary fibroblast cultures. The present paper critically reviews recent data on the potency of somatic stem cells, methods to modify the potency of somatic cells and implications for cell-based therapies.
Collapse
Affiliation(s)
- Wilfried A Kues
- Department of Biotechnology, Institut für Tierzucht, Mariensee, D-31535 Neustadt, Germany.
| | | | | |
Collapse
|
22
|
Anger M, Klima J, Kubelka M, Prochazka R, Motlik J, Schultz RM. Timing of Plk1 and MPF activation during porcine oocyte maturation. Mol Reprod Dev 2004; 69:11-6. [PMID: 15278898 DOI: 10.1002/mrd.20151] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A Polo-like kinase 1 (Plk1) appears involved in an autocatalytic loop between CDC25C phosphatase and M phase promoting factor (MPF) in Xenopus oocytes and leads to activation of MPF that is required for germinal vesicle breakdown (GVBD). Although similar evidence for such a role of Plk1 in MPF activation during maturation of mammalian oocytes is absent, changes in Plk1 enzyme activity correlate with MPF activation, Plk1 co-localizes with MPF, and microinjection of antibodies neutralizing Plk1 delays GVBD. In this study, we exploited the prolonged time required for maturation of porcine oocytes to define precisely the timing of Plk1 and MPF activation during maturation. GVBD typically occurs between 24 and 26 hr of culture in vitro and meiotic maturation is completed after 40-44-hr culture. We find that Plk1 is activated before MPF, which is consistent with its role in activating MPF in mammalian oocytes.
Collapse
Affiliation(s)
- Martin Anger
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104-6018, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Microinjection of foreign DNA into pronuclei of a fertilized oocyte has predominantly been used for the generation of transgenic livestock. This technology works reliably, but is inefficient and results in random integration and variable expression patterns in the transgenic offspring. Nevertheless, remarkable achievements have been made with this technology. By targeting expression to the mammary gland, numerous heterologous recombinant human proteins have been produced in large amounts which could be purified from milk of transgenic goats, sheep, cattle and rabbit. Products such as human anti-thrombin III, alpha-anti-trypsin and tissue plasminogen activator are currently in advanced clinical trials and are expected to be on the market within the next few years. Transgenic pigs that express human complement regulating proteins have been tested in their ability to serve as donors in human organ transplantation (i.e. xenotransplantation). In vitro and in vivo data convincingly show that the hyperacute rejection response can be overcome in a clinically acceptable manner by successful employing this strategy. It is anticipated that transgenic pigs will be available as donors for functional xenografts within a few years. Similarly, pigs may serve as donors for a variety of xenogenic cells and tissues. The recent developments in nuclear transfer and its merger with the growing genomic data allow a targeted and regulatable transgenic production. Systems for efficient homologous recombination in somatic cells are being developed and the adaptation of sophisticated molecular tools, already explored in mice, for transgenic livestock production is underway. The availability of these technologies are essential to maintain "genetic security" and to ensure absence of unwanted side effects.
Collapse
Affiliation(s)
- Heiner Niemann
- Department of Biotechnology, Institut für Tierzucht Mariensee, FAL, 31535 Neustadt, Germany.
| | | |
Collapse
|