1
|
McKay LK, White JP. The AMPK/p27 Kip1 Pathway as a Novel Target to Promote Autophagy and Resilience in Aged Cells. Cells 2021; 10:cells10061430. [PMID: 34201101 PMCID: PMC8229180 DOI: 10.3390/cells10061430] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 11/16/2022] Open
Abstract
Once believed to solely function as a cyclin-dependent kinase inhibitor, p27Kip1 is now emerging as a critical mediator of autophagy, cytoskeletal dynamics, cell migration and apoptosis. During periods of metabolic stress, the subcellular location of p27Kip1 largely dictates its function. Cytoplasmic p27Kip1 has been found to be promote cellular resilience through autophagy and anti-apoptotic mechanisms. Nuclear p27Kip1, however, inhibits cell cycle progression and makes the cell susceptible to quiescence, apoptosis, and/or senescence. Cellular location of p27Kip1 is regulated, in part, by phosphorylation by various kinases, including Akt and AMPK. Aging promotes nuclear localization of p27Kip1 and a predisposition to senescence or apoptosis. Here, we will review the role of p27Kip1 in healthy and aging cells with a particular emphasis on the interplay between autophagy and apoptosis.
Collapse
Affiliation(s)
- Lauren K. McKay
- Adams School of Dentistry, UNC Chapel Hill, Chapel Hill, NC 27599, USA;
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 N. Duke Street, Durham, NC 27701, USA
| | - James P. White
- Duke Molecular Physiology Institute, Duke University School of Medicine, 300 N. Duke Street, Durham, NC 27701, USA
- Department of Medicine, Duke University School of Medicine, 300 N. Duke Street, Durham, NC 27701, USA
- Duke Center for the Study of Aging and Human Development, Duke University School of Medicine, 300 N. Duke Street, Durham, NC 27701, USA
- Correspondence:
| |
Collapse
|
2
|
Bencivenga D, Stampone E, Aulitto A, Tramontano A, Barone C, Negri A, Roberti D, Perrotta S, Della Ragione F, Borriello A. A cancer-associated CDKN1B mutation induces p27 phosphorylation on a novel residue: a new mechanism for tumor suppressor loss-of-function. Mol Oncol 2021; 15:915-941. [PMID: 33316141 PMCID: PMC8024736 DOI: 10.1002/1878-0261.12881] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/16/2020] [Accepted: 12/11/2020] [Indexed: 01/12/2023] Open
Abstract
CDKN1B haploinsufficiency promotes the development of several human cancers. The gene encodes p27Kip1, a protein playing pivotal roles in the control of growth, differentiation, cytoskeleton dynamics, and cytokinesis. CDKN1B haploinsufficiency has been associated with chromosomal or gene aberrations. However, very few data exist on the mechanisms by which CDKN1B missense mutations facilitate carcinogenesis. Here, we report a functional study on a cancer‐associated germinal p27Kip1 variant, namely glycine9‐>arginine‐p27Kip1 (G9R‐p27Kip1) identified in a parathyroid adenoma. We unexpectedly found that G9R‐p27Kip1 lacks the major tumor suppressor activities of p27Kip1 including its antiproliferative and pro‐apoptotic functions. In addition, G9R‐p27Kip1 transfection in cell lines induces the formation of more numerous and larger spheres when compared to wild‐type p27Kip1‐transfected cells. We demonstrated that the mutation creates a consensus sequence for basophilic kinases causing a massive phosphorylation of G9R‐p27Kip1 on S12, a residue normally never found modified in p27Kip1. The novel S12 phosphorylation appears responsible for the loss of function of G9R‐p27Kip1 since S12AG9R‐p27Kip1 recovers most of the p27Kip1 tumor suppressor activities. In addition, the expression of the phosphomimetic S12D‐p27Kip1 recapitulates G9R‐p27Kip1 properties. Mechanistically, S12 phosphorylation enhances the nuclear localization of the mutant protein and also reduces its cyclin‐dependent kinase (CDK)2/CDK1 inhibition activity. To our knowledge, this is the first reported case of quantitative phosphorylation of a p27Kip1 variant on a physiologically unmodified residue associated with the loss of several tumor suppressor activities. In addition, our findings demonstrate that haploinsufficiency might be due to unpredictable post‐translational modifications due to generation of novel consensus sequences by cancer‐associated missense mutations.
Collapse
Affiliation(s)
- Debora Bencivenga
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Emanuela Stampone
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Arianna Aulitto
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annunziata Tramontano
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Clementina Barone
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Aide Negri
- Department of Medicine and Surgery, University of Parma, Italy
| | - Domenico Roberti
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Silverio Perrotta
- Department of Woman, Child and General and Specialized Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Fulvio Della Ragione
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Adriana Borriello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
3
|
Russo GL, Stampone E, Cervellera C, Borriello A. Regulation of p27 Kip1 and p57 Kip2 Functions by Natural Polyphenols. Biomolecules 2020; 10:biom10091316. [PMID: 32933137 PMCID: PMC7564754 DOI: 10.3390/biom10091316] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/01/2020] [Accepted: 09/09/2020] [Indexed: 12/14/2022] Open
Abstract
In numerous instances, the fate of a single cell not only represents its peculiar outcome but also contributes to the overall status of an organism. In turn, the cell division cycle and its control strongly influence cell destiny, playing a critical role in targeting it towards a specific phenotype. Several factors participate in the control of growth, and among them, p27Kip1 and p57Kip2, two proteins modulating various transitions of the cell cycle, appear to play key functions. In this review, the major features of p27 and p57 will be described, focusing, in particular, on their recently identified roles not directly correlated with cell cycle modulation. Then, their possible roles as molecular effectors of polyphenols’ activities will be discussed. Polyphenols represent a large family of natural bioactive molecules that have been demonstrated to exhibit promising protective activities against several human diseases. Their use has also been proposed in association with classical therapies for improving their clinical effects and for diminishing their negative side activities. The importance of p27Kip1 and p57Kip2 in polyphenols’ cellular effects will be discussed with the aim of identifying novel therapeutic strategies for the treatment of important human diseases, such as cancers, characterized by an altered control of growth.
Collapse
Affiliation(s)
- Gian Luigi Russo
- National Research Council, Institute of Food Sciences, 83100 Avellino, Italy;
- Correspondence: (G.L.R.); (A.B.); Tel.: +39-0825-299-331 (G.L.R.)
| | - Emanuela Stampone
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 81031 Napoli, Italy;
| | - Carmen Cervellera
- National Research Council, Institute of Food Sciences, 83100 Avellino, Italy;
| | - Adriana Borriello
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 81031 Napoli, Italy;
- Correspondence: (G.L.R.); (A.B.); Tel.: +39-0825-299-331 (G.L.R.)
| |
Collapse
|
4
|
Rai KR, Chen B, Zhao Z, Chen Y, Hu J, Liu S, Maarouf M, Li Y, Xiao M, Liao Y, Chen JL. Robust expression of p27Kip1 induced by viral infection is critical for antiviral innate immunity. Cell Microbiol 2020; 22:e13242. [PMID: 32596986 DOI: 10.1111/cmi.13242] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/13/2020] [Accepted: 06/24/2020] [Indexed: 12/27/2022]
Abstract
Influenza A virus (IAV) infection regulates the expression of numerous host genes. However, the precise mechanism underlying implication of these genes in IAV pathogenesis remains largely unknown. Here, we employed isobaric tags for relative and absolute quantification (iTRAQ) to identify host proteins regulated by IAV infection. iTRAQ analysis of mouse lungs infected or uninfected with IAV showed a total of 167 differentially upregulated proteins in response to the viral infection. Interestingly, we observed that p27Kip1, a potent cyclin-dependent kinase inhibitor, was markedly induced by IAV both at mRNA and protein levels through in vitro and in vivo studies. Furthermore, it was shown that innate immune signalling positively regulated p27Kip1 expression in response to IAV infection. Ectopic expression of p27Kip1 in A549 cells dramatically inhibited IAV replication, whereas, p27Kip1 knockdown significantly enhanced the virus replication. in vivo experiments demonstrated that p27Kip1 knockout (KO) mice were more susceptible to IAV than wild-type (WT) mice: exhibiting higher viral load in lung tissue, faster body-weight loss, reduced survival rate and more severe organ damage. Moreover, we found that p27Kip1 overexpression facilitated the degradation of viral NS1 protein, caused a dramatic STAT1 activation and promoted the expression of IFN-β and several critical antiviral interferon-stimulated genes (ISGs). Increased p27Kip1 expression also restricted infections of several other viruses. Conversely, IAV-infected p27Kip1 KO mice exhibited a sharp increase in NS1 protein accumulation, reduced level of STAT1 activation and decreased expression of IFN-β and the ISGs in the lung compared to WT animals. These findings reveal a key role of p27Kip1 in enhancing antiviral innate immunity.
Collapse
Affiliation(s)
- Kul Raj Rai
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Biao Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Zhonghui Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuhai Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiayue Hu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shasha Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Mohamed Maarouf
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yingying Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Meng Xiao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Liao
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Ji-Long Chen
- Key Laboratory of Fujian-Taiwan Animal Pathogen Biology, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
5
|
Yumimoto K, Yamauchi Y, Nakayama KI. F-Box Proteins and Cancer. Cancers (Basel) 2020; 12:cancers12051249. [PMID: 32429232 PMCID: PMC7281081 DOI: 10.3390/cancers12051249] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/20/2022] Open
Abstract
Controlled protein degradation is essential for the operation of a variety of cellular processes including cell division, growth, and differentiation. Identification of the relations between ubiquitin ligases and their substrates is key to understanding the molecular basis of cancer development and to the discovery of novel targets for cancer therapeutics. F-box proteins function as the substrate recognition subunits of S-phase kinase-associated protein 1 (SKP1)−Cullin1 (CUL1)−F-box protein (SCF) ubiquitin ligase complexes. Here, we summarize the roles of specific F-box proteins that have been shown to function as tumor promoters or suppressors. We also highlight proto-oncoproteins that are targeted for ubiquitylation by multiple F-box proteins, and discuss how these F-box proteins are deployed to regulate their cognate substrates in various situations.
Collapse
|
6
|
Caglar HO, Biray Avci C. Alterations of cell cycle genes in cancer: unmasking the role of cancer stem cells. Mol Biol Rep 2020; 47:3065-3076. [DOI: 10.1007/s11033-020-05341-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/22/2020] [Indexed: 02/07/2023]
|
7
|
Luo Y, Fu Z, Wu P, Zheng D, Zhang X. The clinicopathological and prognostic significance of P27 kip in hepatocellular carcinoma patients: A systemic review and meta-analysis. Gene 2020; 734:144351. [PMID: 31982553 DOI: 10.1016/j.gene.2020.144351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 01/27/2023]
Abstract
OBJECTIVES P27kip is a cyclin-dependent kinase inhibitor that has gained importance as a biomarker in human malignant tumors. However, the potential role of P27kip in hepatocellular carcinoma remains unclear. The aim of this meta-analysis was to explore whether P27kip acts as prognostic and clinicopathological roles in hepatocellular carcinoma patients. METHODS/MATERIALS An electronic search based on three databases, PubMed, Embase, and Web of Science, was performed to select a sufficient number of studies. Pooled hazard ratio (HR) and odds ratio (OR) were used as estimates to investigate the association among P27kip expression, prognosis and clinicopathological features. RESULTS In total, we identified 18 studies with 1774 hepatocellular carcinoma patients. The result derived from four studies revealed a significant positive association between lower P27kip levels and shorter overall survival (HR = 0.550, 95% CI: 0.464-0.652, P < 0001) and disease-free survival (HR = 0.420, 95% CI: 0.308-0.571, P < 0.0001). Analyses of the clinicopathological features and P27kip expression also showed that a positive rate of P27kip was significantly lower in a larger sized tumor (OR = 0.538, 95% CI: 0.315-0.919, P = 0.023). The results also revealed that lower P27kip levels were correlated with poorer differentiation (0.416, 95% CI: 0.178-0.971, P = 0.043). Additionally, the pooled OR of 0.389 also presented a significant correlation between P27kip underexpression and the metastasis of HCCs (95% CI: 0.155-0.975, P = 0.044). CONCLUSIONS This analysis suggests a strong association among P27kip underexpression, poorer prognosis and aggressive progression of hepatocellular carcinoma in patients. P27kip may be a tumor suppressor for predicting the progression and survival outcome in patients with hepatocellular carcinomas.
Collapse
Affiliation(s)
- Yongxiang Luo
- Department of General Surgery Second Ward, The First People's Hospital of Qinzhou, Qinzhou, Guangxi Zhuang Autonomous Region 535000, People's Republic of China
| | - Zhaoyin Fu
- Department of Intensive Care Unit, The First People's Hospital of Qinzhou, Qinzhou, Guangxi Zhuang Autonomous Region 535000, People's Republic of China
| | - Peisheng Wu
- Department of General Surgery Second Ward, The First People's Hospital of Qinzhou, Qinzhou, Guangxi Zhuang Autonomous Region 535000, People's Republic of China
| | - Dawu Zheng
- Department of General Surgery Second Ward, The First People's Hospital of Qinzhou, Qinzhou, Guangxi Zhuang Autonomous Region 535000, People's Republic of China.
| | - Xiumei Zhang
- Department of General Surgery Second Ward, The First People's Hospital of Qinzhou, Qinzhou, Guangxi Zhuang Autonomous Region 535000, People's Republic of China
| |
Collapse
|
8
|
Shi H, Li H, Yuan R, Guan W, Zhang X, Zhang S, Zhang W, Tong F, Li L, Song Z, Wang C, Yang S, Wang H. PCBP1 depletion promotes tumorigenesis through attenuation of p27 Kip1 mRNA stability and translation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:187. [PMID: 30086790 PMCID: PMC6081911 DOI: 10.1186/s13046-018-0840-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 07/10/2018] [Indexed: 11/10/2022]
Abstract
Background Poly C Binding Protein 1 (PCBP1) is an RNA-binding protein that binds and regulates translational activity of subsets of cellular mRNAs. Depletion of PCBP1 is implicated in various carcinomas, but the underlying mechanism in tumorigenesis remains elusive. Methods We performed a transcriptome-wide screen to identify novel bounding mRNA of PCBP1. The bind regions between PCBP1 with target mRNA were investigated by using point mutation and luciferase assay. Cell proliferation, cell cycle, tumorigenesis and cell apoptosis were also evaluated in ovary and colon cancer cell lines. The mechanism that PCBP1 affects p27 was analyzed by mRNA stability and ribosome profiling assays. We analyzed PCBP1 and p27 expression in ovary, colon and renal tumor samples and adjacent non-tumor tissues using RT-PCR, Western Blotting and immunohistochemistry. The prognostic significance of PCBP1 and p27 also analyzed using online databases. Results We identified cell cycle inhibitor p27Kip1 (p27) as a novel PCBP1-bound transcript. We then demonstrated that binding of PCBP1 to p27 3’UTR via its KH1 domain mainly stabilizes p27 mRNA, while enhances its translation to fuel p27 expression, prior to p27 protein degradation. The upregulated p27 consequently inhibits cell proliferation, cell cycle progression and tumorigenesis, whereas promotes cell apoptosis under paclitaxel treatment. Conversely, knockdown of PCBP1 in turn compromises p27 mRNA stability, leading to lower p27 level and tumorigenesis in vivo. Moreover, forced depletion of p27 counteracts the tumor suppressive ability of PCBP1 in the same PCBP1 over-expressing cells. Physiologically, we showed that decreases of both p27 mRNA and its protein expressions are well correlated to PCBP1 depletion in ovary, colon and renal tumor samples, independent of the p27 ubiquitin ligase Skp2 level. Correlation of PCBP1 with p27 is also found in the tamoxifen, doxorubincin and lapatinib resistant breast cancer cells of GEO database. Conclusion Our results thereby indicate that loss of PCBP1 expression firstly attenuates p27 expression at post-transcriptional level, and subsequently promotes carcinogenesis. PCBP1 could be used as a diagnostic marker to cancer patients. Electronic supplementary material The online version of this article (10.1186/s13046-018-0840-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hongshun Shi
- Centre for Translational Medicine, the First Affiliated Hospital, SUN Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.,Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Hui Li
- Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China.,Center for Stem Cell Biology and Tissue Engineering, Key laboratory of ministry of education, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Ronghua Yuan
- Department of General Surgery, The Second Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
| | - Wen Guan
- Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Xiaomei Zhang
- Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Shaoyang Zhang
- Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Wenliang Zhang
- Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Fang Tong
- Centre for Translational Medicine, the First Affiliated Hospital, SUN Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.,Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Li Li
- Centre for Translational Medicine, the First Affiliated Hospital, SUN Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.,Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Zhihong Song
- Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China
| | - Changwei Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Shulan Yang
- Centre for Translational Medicine, the First Affiliated Hospital, SUN Yat-sen University, 58 Second Zhongshan Road, Guangzhou, 510080, China.
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, SUN Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China. .,Center for Stem Cell Biology and Tissue Engineering, Key laboratory of ministry of education, Sun Yat-sen University, 74 Second Zhongshan Road, Guangzhou, 510080, China.
| |
Collapse
|
9
|
Bencivenga D, Caldarelli I, Stampone E, Mancini FP, Balestrieri ML, Della Ragione F, Borriello A. p27 Kip1 and human cancers: A reappraisal of a still enigmatic protein. Cancer Lett 2017; 403:354-365. [DOI: 10.1016/j.canlet.2017.06.031] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 12/21/2022]
|
10
|
Oi N, Yamamoto H, Langfald A, Bai R, Lee MH, Bode AM, Dong Z. LTA4H regulates cell cycle and skin carcinogenesis. Carcinogenesis 2017; 38:728-737. [PMID: 28575166 PMCID: PMC6248358 DOI: 10.1093/carcin/bgx049] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/11/2017] [Accepted: 05/24/2017] [Indexed: 12/30/2022] Open
Abstract
Leukotriene A4 hydrolase (LTA4H), a bifunctional zinc metallo-enzyme, is reportedly overexpressed in several human cancers. Our group has focused on LTA4H as a potential target for cancer prevention and/or therapy. In the present study, we report that LTA4H is a key regulator of cell cycle at the G0/G1 phase acting by negatively regulating p27 expression in skin cancer. We found that LTA4H is overexpressed in human skin cancer tissue. Knocking out LTA4H significantly reduced skin cancer development in the 7,12-dimethylbenz(a)anthracene (DMBA)-initiated/12-O-tetradecanoylphorbol-13-acetate (TPA)-promoted two-stage skin cancer mouse model. LTA4H depletion dramatically decreased anchorage-dependent and -independent skin cancer cell growth by inducing cell cycle arrest at the G0/G1 phase. Moreover, our findings showed that depletion of LTA4H enhanced p27 protein stability, which was associated with decreased phosphorylation of CDK2 at Thr160 and inhibition of the CDK2/cyclin E complex, resulting in down-regulated p27 ubiquitination. These findings indicate that LTA4H is critical for skin carcinogenesis and is an important mediator of cell cycle and the data begin to clarify the mechanisms of LTA4H's role in cancer development.
Collapse
Affiliation(s)
- Naomi Oi
- The Hormel Institute, University of Minnesota, 801 16th Ave. NE, Austin, MN 55912, USA
| | - Hiroyuki Yamamoto
- The Hormel Institute, University of Minnesota, 801 16th Ave. NE, Austin, MN 55912, USA
| | - Alyssa Langfald
- The Hormel Institute, University of Minnesota, 801 16th Ave. NE, Austin, MN 55912, USA
| | - Ruihua Bai
- The Hormel Institute, University of Minnesota, 801 16th Ave. NE, Austin, MN 55912, USA
| | - Mee-Hyun Lee
- The Hormel Institute, University of Minnesota, 801 16th Ave. NE, Austin, MN 55912, USA
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, 801 16th Ave. NE, Austin, MN 55912, USA
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, 801 16th Ave. NE, Austin, MN 55912, USA
| |
Collapse
|
11
|
Lang MA, Jenkins SA, Balzano P, Owoyele A, Patel A, Bamezai AK. Engaging Ly-6A/Sca-1 triggers lipid raft-dependent and -independent responses in CD4 + T-cell lines. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:448-460. [PMID: 28660664 PMCID: PMC5691314 DOI: 10.1002/iid3.182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 05/22/2017] [Accepted: 05/26/2017] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The lymphocyte antigen 6 (Ly-6) supergene family encodes proteins of 12-14 kda in molecular mass that are either secreted or anchored to the plasma membrane through a glycosyl-phosphatidylinisotol (GPI) lipid anchor at the carboxy-terminus. The lipidated GPI-anchor allows localization of Ly-6 proteins to the 10-100 nm cholesterol-rich nano-domains on the membrane, also known as lipid rafts. Ly-6A/Sca-1, a member of Ly-6 gene family is known to transduce signals despite the absence of transmembrane and cytoplasmic domains. It is hypothesized that the localization of Ly-6A/Sca-1 with in lipid rafts allows this protein to transduce signals to the cell interior. METHODS AND RESULTS In this study, we found that cross-linking mouse Ly-6A/Sca-1 protein with a monoclonal antibody results in functionally distinct responses that occur simultaneously. Ly-6A/Sca-1 triggered a cell stimulatory response as gauged by cytokine production with a concurrent inhibitory response as indicated by growth inhibition and apoptosis. While production of interleukin 2 (IL-2) cytokine by CD4+ T cell line in response to cross-linking Ly-6A/Sca-1 was dependent on the integrity of lipid rafts, the observed cell death occurred independently of it. Growth inhibited CD4+ T cells showed up-regulated expression of the inhibitory cell cycle protein p27kip but not of p53. In addition, Ly-6A/Sca-1 induced translocation of cytochrome C to the cytoplasm along with activated caspase 3 and caspase 9, thereby suggesting an intrinsic apoptotic cell death mechanism. CONCLUSIONS We conclude that opposing responses with differential dependence on the integrity of lipid rafts are triggered by engaging Ly-6A/Sca-1 protein on the membrane of transformed CD4+ T cells.
Collapse
Affiliation(s)
- Melissa A Lang
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Sultan A Jenkins
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Phillip Balzano
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Adeyinka Owoyele
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Akshay Patel
- Department of Biology, Villanova University, Villanova, Pennsylvania
| | - Anil K Bamezai
- Department of Biology, Villanova University, Villanova, Pennsylvania
| |
Collapse
|
12
|
Hammerich KH, Frolov A, Li R, Ittmann M, Ayala GE. Cellular interactions of the phosphorylated form of AKT in prostate cancer. Hum Pathol 2017; 63:98-109. [DOI: 10.1016/j.humpath.2017.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/25/2017] [Accepted: 02/23/2017] [Indexed: 11/15/2022]
|
13
|
Sharma SS, Ma L, Pledger WJ. p27Kip1 inhibits the cell cycle through non-canonical G1/S phase-specific gatekeeper mechanism. Cell Cycle 2016; 14:3954-64. [PMID: 26697844 DOI: 10.1080/15384101.2015.1100775] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The cyclin-dependent kinase (CDK) inhibitor p27Kip1 has been shown to regulate cellular proliferation via inhibition of CDK activities. It is now recognized that p27Kip1 can regulate cellular processes through non-canonical, CDK-independent mechanisms. We have developed an inducible p27Kip1 model in cultured cells to explore CDK-independent p27Kip1 regulation of biological processes. We present evidence that p27Kip1 can function in a CDK-independent manner to inhibit entry and/or progression of S phase. Even though this p27Kip1 mechanism is non-canonical it does requires the intact cyclin-binding motif in p27Kip1. We suggest a mechanism similar to that proposed in post-mitotic neural cells whereby p27Kip1 functions to coordinate growth arrest and apoptosis. Our hypothesis supports the concept that p27Kip1 is a gatekeeper for the entry and progression of S phase through interaction with specific protein(s) or via binding to specific DNA sequences in a CDK-independent manner.
Collapse
Affiliation(s)
| | - Le Ma
- a Gibbs Cancer Center and Research Institute ; Spartanburg , SC
| | - W Jackson Pledger
- a Gibbs Cancer Center and Research Institute ; Spartanburg , SC.,b Edward Via College of Osteopathic Medicine ; Department of Molecular Medicine ; Spartanburg , SC USA
| |
Collapse
|
14
|
Seo HH, Lee CY, Lee J, Lim S, Choi E, Park JC, Lee S, Hwang KC. The role of nuclear factor of activated T cells during phorbol myristate acetate-induced cardiac differentiation of mesenchymal stem cells. Stem Cell Res Ther 2016; 7:90. [PMID: 27405982 PMCID: PMC4942985 DOI: 10.1186/s13287-016-0348-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 06/03/2016] [Accepted: 06/17/2016] [Indexed: 11/16/2022] Open
Abstract
Background We previously reported that phorbol 12-myristate 13-acetate (PMA) treatment can induce the cardiac differentiation of mesenchymal stem cells (MSCs). In the present study, we investigated how PMA induces cardiac differentiation of MSCs, focusing on its effect on the transcription factors responsible for increased cardiac marker gene expression. Methods Human MSCs (hMSCs) were treated with 1 μM PMA for 9 days. The expression of MSC markers and cardiac markers in the PMA-treated hMSC, as well as the nuclear translocation of transcription factors, nuclear factor of activated T cells (NFAT), and myogenic differentiation 1 (MyoD), was examined. Transcriptional activity of NFAT was examined by utilizing a green fluorescent protein (GFP) vector containing NFAT motif of human interleukin-2 promoter. The effect of PMA on the expression of key cell cycle regulators was examined. Results PMA induces the transcriptional activity of NFAT and MyoD, which have been associated with increased expression of cardiac troponin T (cTnT) and myosin heavy chain (MHC), respectively. Our data suggested that protein kinase C (PKC) mediates the effect of PMA on NFAT activation. Furthermore, PMA treatment increased cell-cycle regulator p27kip1 expression, suggesting that PMA triggers the cardiac differentiation program in MSCs by regulating key transcription factors and cell cycle regulators. Conclusions The results of this study demonstrate the importance of NFAT activation during PMA-induced MSC differentiation and help us to better understand the underlying mechanisms of small molecule-mediated MSC differentiation so that we can develop a strategy for synthesizing novel and improved differentiation-inducing small molecules. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0348-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea
| | - Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, Seoul, South Korea
| | - Jiyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, South Korea
| | - Soyeon Lim
- Institute for Bio-medical Convergence, Catholic Kwandong University, Incheon, South Korea
| | - Eunhyun Choi
- Institute for Bio-medical Convergence, Catholic Kwandong University, Incheon, South Korea
| | - Jong-Chul Park
- Cellbiocontrol Laboratory, Department of Medical Engineering, Yonsei University College of Medicine, Seoul, South Korea
| | - Seahyoung Lee
- Institute for Bio-medical Convergence, Catholic Kwandong University, Incheon, South Korea. .,Department of Biomedical Sciences, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, South Korea.
| | - Ki-Chul Hwang
- Institute for Bio-medical Convergence, Catholic Kwandong University, Incheon, South Korea. .,Department of Biomedical Sciences, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, South Korea.
| |
Collapse
|
15
|
Abstract
The preimplantation development stage of mammalian embryogenesis consists of a series of highly conserved, regulated, and predictable cell divisions. This process is essential to allow the rapid expansion and differentiation of a single-cell zygote into a multicellular blastocyst containing cells of multiple developmental lineages. This period of development, also known as the germinal stage, encompasses several important developmental transitions, which are accompanied by dramatic changes in cell cycle profiles and dynamics. These changes are driven primarily by differences in the establishment and enforcement of cell cycle checkpoints, which must be bypassed to facilitate the completion of essential cell cycle events. Much of the current knowledge in this area has been amassed through the study of knockout models in mice. These mouse models are powerful experimental tools, which have allowed us to dissect the relative dependence of the early embryonic cell cycles on various aspects of the cell cycle machinery and highlight the extent of functional redundancy between members of the same gene family. This chapter will explore the ways in which the cell cycle machinery, their accessory proteins, and their stimuli operate during mammalian preimplantation using mouse models as a reference and how this allows for the usually well-defined stages of the cell cycle to be shaped and transformed during this unique and critical stage of development.
Collapse
|
16
|
Abstract
p27(Kip1) was first discovered as a key regulator of cell proliferation. The canonical function of p27(Kip1) is inhibition of cyclin-dependent kinase (CDK) activity. In addition to its initial identification as a CDK inhibitor, p27(Kip1) has also emerged as an intrinsically unstructured, multifunctional protein with numerous non-canonical, CDK-independent functions that exert influence on key processes such as cell cycle regulation, cytoskeletal dynamics and cellular plasticity, cell migration, and stem-cell proliferation and differentiation. Many of these non-canonical functions, depending on the cell-specific contexts such as oncogenic activation of signaling pathways, have the ability to turn pro-oncogenic in nature and even contribute to tumor-aggressiveness and metastasis. This review discusses the various non-canonical, CDK-independent mechanisms by which p27(Kip1) functions either as a tumor-suppressor or tumor-promoter.
Collapse
Affiliation(s)
- Savitha S Sharma
- a Gibbs Cancer Center & Research Institute , Spartanburg , SC , USA
| | - W Jackson Pledger
- a Gibbs Cancer Center & Research Institute , Spartanburg , SC , USA.,b Edward Via College of Osteopathic Medicine , Department of Molecular Medicine , Spartanburg , SC , USA
| |
Collapse
|
17
|
Podmirseg SR, Jäkel H, Ranches GD, Kullmann MK, Sohm B, Villunger A, Lindner H, Hengst L. Caspases uncouple p27(Kip1) from cell cycle regulated degradation and abolish its ability to stimulate cell migration and invasion. Oncogene 2016; 35:4580-90. [PMID: 26829051 PMCID: PMC4854979 DOI: 10.1038/onc.2015.524] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 10/27/2015] [Accepted: 11/06/2015] [Indexed: 01/12/2023]
Abstract
In addition to their role in programmed cell death, caspases exert non-lethal functions in diverse developmental processes including cell differentiation or tissue remodeling. Terminal cell cycle exit and differentiation can be promoted by increased level of the CDK inhibitor p27Kip1. Activated caspases cause proteolytic processing of p27, and we identified a novel caspase cleavage site in human p27 that removes a C-terminal fragment of 22 amino acids from the CDK inhibitor, including a phosphodegron. Thereby, caspases protect the inhibitor from SCF-Skp2-mediated degradation in S, G2 and M phases of the cell cycle. As a consequence, p27 becomes stabilized and remains an efficient nuclear inhibitor of cell cycle progression. Besides controlling cyclin/CDK kinase activity, p27 also regulates cytoskeletal dynamics, cell motility and cell invasion. Following processing by caspases, p27 fails to bind to RhoA and to inhibit its activation, and thereby abolishes the ability of p27 to stimulate cell migration and invasion. We propose that the stabilization of the CDK inhibitor and elimination of RhoA-induced cytoskeletal remodeling upon caspase processing could contribute to cell cycle exit and cytoskeletal remodeling during non-lethal caspase controlled differentiation processes.
Collapse
Affiliation(s)
- S R Podmirseg
- Division of Medical Biochemistry; Biocenter; Innsbruck Medical University; Innsbruck, Austria
| | - H Jäkel
- Division of Medical Biochemistry; Biocenter; Innsbruck Medical University; Innsbruck, Austria
| | - G D Ranches
- Division of Medical Biochemistry; Biocenter; Innsbruck Medical University; Innsbruck, Austria
| | - M K Kullmann
- Division of Medical Biochemistry; Biocenter; Innsbruck Medical University; Innsbruck, Austria
| | - B Sohm
- Laboratoire Interdisciplinaire des Environnements Continentaux (LIEC), UMR 7360, Université de Lorraine, Metz, France.,CNRS, LIEC, UMR 7360, Metz, France
| | - A Villunger
- Division of Developmental Immunology; Biocenter; Innsbruck Medical University; Innsbruck, Austria.,Tyrolean Cancer Research Institute, Innsbruck, Austria
| | - H Lindner
- Division of Clinical Biochemistry; Biocenter; Innsbruck Medical University; Innsbruck, Austria
| | - L Hengst
- Division of Medical Biochemistry; Biocenter; Innsbruck Medical University; Innsbruck, Austria
| |
Collapse
|
18
|
Lovisa S, Citro S, Sonego M, Dall'Acqua A, Ranzuglia V, Berton S, Colombatti A, Belletti B, Chiocca S, Schiappacassi M, Baldassarre G. SUMOylation regulates p27Kip1 stability and localization in response to TGFβ. J Mol Cell Biol 2015; 8:17-30. [PMID: 26450989 DOI: 10.1093/jmcb/mjv056] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 06/08/2015] [Indexed: 11/13/2022] Open
Abstract
Exposure of normal and tumor-derived cells to TGFβ results in different outcomes, depending on the regulation of key targets. The CDK inhibitor p27(Kip1) is one of these TGFβ targets and is essential for the TGFβ-induced cell cycle arrest. TGFβ treatment inhibits p27(Kip1) degradation and induces its nuclear translocation, through mechanisms that are still unknown. Recent evidences suggest that SUMOylation, a post-translational modification able to modulate the stability and subcellular localization of target proteins, critically modifies members of the TGFβ signaling pathway. Here, we demonstrate that p27(Kip1) is SUMOylated in response to TGFβ treatment. Using different p27(Kip1) point mutants, we identified lysine 134 (K134) as the residue modified by small ubiquitin-like modifier 1 (SUMO1) in response to TGFβ treatment. TGFβ-induced K134 SUMOylation increased protein stability and nuclear localization of both endogenous and exogenously expressed p27(Kip1). We observed that SUMOylation regulated p27(Kip1) binding to CDK2, thereby governing its nuclear proteasomal degradation through the phosphorylation of threonine 187. Importantly, p27(Kip1) SUMOylation was necessary for proper cell cycle exit following TGFβ treatment. These data indicate that SUMOylation is a novel regulatory mechanism that modulates p27(Kip1) function in response to TGFβ stimulation. Given the involvement of TGFβ signaling in cancer cell proliferation and invasion, our data may shed light on an important aspect of this pathway during tumor progression.
Collapse
Affiliation(s)
- Sara Lovisa
- Division of Experimental Oncology 2 Centro di Riferimento Oncologico, National Cancer Institute, Aviano 33081, Italy
| | - Simona Citro
- Department of Experimental Oncology, European Institute of Oncology at the IFOM-IEO Campus, Milan 20139, Italy
| | - Maura Sonego
- Division of Experimental Oncology 2 Centro di Riferimento Oncologico, National Cancer Institute, Aviano 33081, Italy
| | - Alessandra Dall'Acqua
- Division of Experimental Oncology 2 Centro di Riferimento Oncologico, National Cancer Institute, Aviano 33081, Italy
| | - Valentina Ranzuglia
- Division of Experimental Oncology 2 Centro di Riferimento Oncologico, National Cancer Institute, Aviano 33081, Italy
| | - Stefania Berton
- Division of Experimental Oncology 2 Centro di Riferimento Oncologico, National Cancer Institute, Aviano 33081, Italy
| | - Alfonso Colombatti
- Division of Experimental Oncology 2 Centro di Riferimento Oncologico, National Cancer Institute, Aviano 33081, Italy Department of Scienze e Tecnologie Biomediche, MATI Center of Excellence, University of Udine, Udine 33010, Italy
| | - Barbara Belletti
- Division of Experimental Oncology 2 Centro di Riferimento Oncologico, National Cancer Institute, Aviano 33081, Italy
| | - Susanna Chiocca
- Department of Experimental Oncology, European Institute of Oncology at the IFOM-IEO Campus, Milan 20139, Italy
| | - Monica Schiappacassi
- Division of Experimental Oncology 2 Centro di Riferimento Oncologico, National Cancer Institute, Aviano 33081, Italy
| | - Gustavo Baldassarre
- Division of Experimental Oncology 2 Centro di Riferimento Oncologico, National Cancer Institute, Aviano 33081, Italy
| |
Collapse
|
19
|
Abstract
PURPOSE P27(kip1) is a negative cell cycle regulator that plays an important role in tumor suppression. Deregulation of p27(kip1) is commonly observed in many human cancers. Numerous studies about p27(kip1) are reported in clinical patients despite variable data for the prognostic of p27(kip1) expression. Here we report a meta-analysis of the association of p27(kip1) expression with the survival of ovarian cancer. METHODS PubMed and Web of science were searched for studies evaluating expression of p27(kip1) and prognostic in ovarian cancer. Published data were extracted and computed into odds ratios (ORs) for death at 3 and 5 years. Data were pooled using the random-effect model. All statistical tests were two-sided. RESULTS Analysis included 9 studies: six studies were reported in European, three studies were reported in American, and one study was reported in Asian. Loss of p27(kip1) was associated with worse overall survival (OS) at both 3 years [OR = 2.61, 95 % confidence interval (CI) 1.95-3.49, p < 0.05] and 5 years (OR = 3.01, 95 % CI 2.17-4.17, p < 0.05). Among studies with different ethnicity (European, American and Asian), the results showed a more significant association in European, including Italy, Germany, and Greece [for both 3-year OS (OR = 3.53, 95 % CI 2.37-5.26) and 5-year OS (OR = 3.66, 95 % CI 2.30-5.83)]. CONCLUSIONS Loss of p27(kip1) is associated with worse survival in ovarian cancer. The development of strategies target p27(kip1) could be a reasonable therapeutic approach.
Collapse
|
20
|
Zhao D, Besser AH, Wander SA, Sun J, Zhou W, Wang B, Ince T, Durante MA, Guo W, Mills G, Theodorescu D, Slingerland J. Cytoplasmic p27 promotes epithelial-mesenchymal transition and tumor metastasis via STAT3-mediated Twist1 upregulation. Oncogene 2015; 34:5447-59. [PMID: 25684140 PMCID: PMC4537852 DOI: 10.1038/onc.2014.473] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Revised: 11/24/2014] [Accepted: 12/19/2014] [Indexed: 12/12/2022]
Abstract
p27 restrains normal cell growth, but PI3K-dependent C-terminal phosphorylation of p27 at threonine 157 (T157) and T198 promotes cancer cell invasion. Here, we describe an oncogenic feedforward loop in which p27pT157pT198 binds Janus kinase 2 (JAK2) promoting STAT3 (signal transducer and activator of transcription 3) recruitment and activation. STAT3 induces TWIST1 to drive a p27-dependent epithelial-mesenchymal transition (EMT) and further activates AKT contributing to acquisition and maintenance of metastatic potential. p27 knockdown in highly metastatic PI3K-activated cells reduces STAT3 binding to the TWIST1 promoter, TWIST1 promoter activity and TWIST1 expression, reverts EMT and impairs metastasis, whereas activated STAT3 rescues p27 knockdown. Cell cycle-defective phosphomimetic p27T157DT198D (p27CK-DD) activates STAT3 to induce a TWIST1-dependent EMT in human mammary epithelial cells and increases breast and bladder cancer invasion and metastasis. Data support a mechanism in which PI3K-deregulated p27 binds JAK2, to drive STAT3 activation and EMT through STAT3-mediated TWIST1 induction. Furthermore, STAT3, once activated, feeds forward to further activate AKT.
Collapse
Affiliation(s)
- D Zhao
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - A H Besser
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - S A Wander
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - J Sun
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Zhou
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - B Wang
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - T Ince
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Pathology, Stem Cell Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - M A Durante
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Guo
- Department of Bioinformatics and Computational Biology, and Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, USA
| | - G Mills
- Department of Bioinformatics and Computational Biology, and Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, USA
| | - D Theodorescu
- University of Colorado Cancer Center, University of Colorado, Aurora, CO, USA
| | - J Slingerland
- Braman Family Breast Cancer Institute at Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.,Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
21
|
Wang HC, Lee WS. Progesterone induces RhoA Inactivation in male rat aortic smooth muscle cells through up-regulation of p27(kip1.). Endocrinology 2014; 155:4473-82. [PMID: 25137028 DOI: 10.1210/en.2014-1344] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previously, we showed that progesterone (P4) at physiologic concentrations (5nM-500nM) inhibits proliferation and migration of rat aortic smooth muscle cells (RASMCs). The P4-induced migration inhibition in RASMC was resulted from Rat sacroma homolog gene family, member A (RhoA) inactivation induced by activating the cSrc/AKT/ERK 2/p38 mitogen-activated protein kinase-mediated signaling pathway. We also demonstrated that up-regulation of cyclin-dependent kinase inhibitor 1B (p27(kip1)) is involved in the P4-induced migration inhibition in RASMC. Because P4 can increase formation of the p27(kip1)-RhoA complex in RASMC, this finding led us to hypothesize that the P4-induced inactivation in RhoA might be caused by up-regulation of p27(kip1). Here, we showed that P4 increased phosphorylation of p27(kip1) at Ser10 in the nucleus, which in turn caused p27(kip1) translocation from the nucleus to the cytosol, subsequently increasing formation of the p27(kip1)-RhoA complex. These effects were blocked by knocking-down kinase-interacting stathmin (KIS) using KIS small interfering RNA. Knock-down of p27(kip1) abolished the P4-induced decreases in the level of RhoA protein in RASMC. However, pretreatment of RASMC with the proteasome inhibitor, N-(benzyloxycarbonyl)leucinylleucinylleucinal (MG132), prevented the P4-induced degradation of p27(kip1) and RhoA. Taken together, our investigation of P4-induced migration inhibition in RASMC showed a sequence of associated intracellular events that included 1) increase in formation of the KIS-p27(kip1) complex in the nucleus; 2) phosphorylated nuclear p27(kip1) at Ser10; 3) increased cytosolic translocation of p27(kip1) and formation of the p27(kip1)-RhoA complex in the cytosol; and 4) degradation of p27(kip1) and RhoA through the ubiquitin-proteasome pathway. These findings highlight the molecular mechanisms underlying P4-induced migration inhibition in RASMC.
Collapse
Affiliation(s)
- Hui-Chen Wang
- Graduate Institute of Medical Sciences (H.-C.W., W.-S.L.) and Department of Physiology (W.-S.L.), College of Medicine, Taipei Medical University, Taipei 110, Taiwan; and Cancer Research Center (W.-S.L.), Taipei Medical University Hospital, Taipei 110, Taiwan
| | | |
Collapse
|
22
|
Fu Y, Liu X, Zhou N, Du L, Sun Y, Zhang X, Ge Y. MicroRNA-200b Stimulates Tumour Growth in TGFBR2-Null Colorectal Cancers by Negatively Regulating p27/kip1. J Cell Physiol 2014; 229:772-82. [DOI: 10.1002/jcp.24497] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 10/16/2013] [Indexed: 12/25/2022]
Affiliation(s)
- Yuxuan Fu
- Department of Physiology; Nanjing Medical University; Nanjing People's Republic of China
| | - Xianghua Liu
- Department of Biochemistry and Molecular Biology; Nanjing Medical University; Nanjing People's Republic of China
| | - Ningtian Zhou
- Department of Cardiology; First Affiliated Hospital of Nanjing Medical University; Nanjing People's Republic of China
| | - Lijian Du
- The Laboratory Center for Basic Medical Sciences; Nanjing Medical University; Nanjing People's Republic of China
| | - Yu Sun
- Department of Orthopedics; Clinical Medical College of Yangzhou University; Subei People's Hospital of Jiangsu Province; Yangzhou People's Republic of China
| | - Xiang Zhang
- Department of Physiology; Nanjing Medical University; Nanjing People's Republic of China
| | - Yingbin Ge
- Department of Physiology; Nanjing Medical University; Nanjing People's Republic of China
| |
Collapse
|
23
|
Bustany S, Cahu J, Moros A, Troussard X, Gaël R, Sola B. Expression and subcellular localization of cyclin-dependent kinase inhibitor p27 does not correlate with proliferation pattern of mantle lymphoma cells. Leuk Lymphoma 2013; 55:2204-6. [PMID: 24354680 DOI: 10.3109/10428194.2013.871633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Sophie Bustany
- Normandie University, UNICAEN, MILPAT (EA 4652) , Caen , France
| | | | | | | | | | | |
Collapse
|
24
|
Ooi LC, Watanabe N, Futamura Y, Sulaiman SF, Darah I, Osada H. Identification of small molecule inhibitors of p27(Kip1) ubiquitination by high-throughput screening. Cancer Sci 2013; 104:1461-7. [PMID: 23910095 DOI: 10.1111/cas.12246] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 07/16/2013] [Accepted: 07/25/2013] [Indexed: 01/04/2023] Open
Abstract
Dysregulation of p27(Kip1) due to proteolysis that involves the ubiquitin ligase (SCF) complex with S-phase kinase-associated protein 2 (Skp2) as the substrate-recognition component (SCF(Skp2)) frequently results in tumorigenesis. In this report, we developed a high-throughput screening system to identify small-molecule inhibitors of p27(Kip1) degradation. This system was established by tagging Skp2 with fluorescent monomeric Azami Green (mAG) and CDK subunit 1 (Cks1) (mAGSkp2-Cks1) to bind to p27(Kip1) phosphopeptides. We identified two compounds that inhibited the interaction between mAGSkp2-Cks1 and p27(Kip1): linichlorin A and gentian violet. Further studies have shown that the compounds inhibit the ubiquitination of p27(Kip1) in vitro as well as p27(Kip1) degradation in HeLa cells. Notably, both compounds exhibited preferential antiproliferative activity against HeLa and tsFT210 cells compared with NIH3T3 cells and delayed the G1 phase progression in tsFT210 cells. Our approach indicates a potential strategy for restoring p27(Kip1) levels in human cancers.
Collapse
Affiliation(s)
- Li-Ching Ooi
- Antibiotics Laboratory, RIKEN, Hirosawa, Wako-shi, Saitama, Japan; School of Biological Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
The transforming growth factor-β (TGF-β) system signals via protein kinase receptors and SMAD mediators to regulate a large number of biological processes. Alterations of the TGF-β signalling pathway are implicated in human cancer. Prior to tumour initiation and early during progression, TGF-β acts as a tumour suppressor; however, at later stages, it is often a tumour promoter. Knowledge about the mechanisms involved in TGF-β signal transduction has allowed a better understanding of cancer progression, invasion, metastasis and epithelial-to-mesenchymal transition. Furthermore, several molecular targets with great potential in therapeutic interventions have been identified. This review discusses the TGF-β signalling pathway, its involvement in cancer and current therapeutic approaches.
Collapse
|
26
|
Osaki LH, Gama P. MAPK signaling pathway regulates p27 phosphorylation at threonin 187 as part of the mechanism triggered by early-weaning to induce cell proliferation in rat gastric mucosa. PLoS One 2013; 8:e66651. [PMID: 23762493 PMCID: PMC3676350 DOI: 10.1371/journal.pone.0066651] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 05/10/2013] [Indexed: 12/12/2022] Open
Abstract
During rat postnatal development, gastric cell proliferation and differentiation depend on many elements, which include dietary pattern, hormones, growth factors and their signaling pathways. Among them, EGFR activity is increased through MAPK and Src cascades in response to early weaning that represents the abrupt transition from milk to solid food. We herein investigated the direct involvement of ERK pathway in the control of cell cycle progression during early weaning, and studied the specific role of p27. At 15 days, Wistar rats were separated from dams, fed with powdered chow and daily injected with PD98059 (MEK inhibitor, 300 µg/kg) or 0.5% DMSO (control). By using HE staining and immunohistochemistry for PCNA, we respectively detected mitotic (MI) and proliferative (PI) indices in 18-day-old pups, and observed that both were reduced by PD98059. As cell cycle-related proteins (cyclin E, CDK2, cyclin D1, CDK4, p21 and p27) are involved in proliferative regulation, we compared samples obtained at 17 days in the morning (17 d) and evening (17.5 d). We found that they were not altered after ERK inhibition, but cyclin D1, p21 and p27 levels changed throughout the day in the control group. As p27 activity depends on its integrity, we studied p27 phosphorylation (threonin 187), and observed that ERK inhibition reduced this process. We suggest that MAPK pathway interferes in the regulation of p27 function in the gastric mucosa during early weaning, possibly by controlling its degradation, and altogether this mechanism might contribute to the increase of epithelial proliferation at this condition.
Collapse
Affiliation(s)
- Luciana H Osaki
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | | |
Collapse
|
27
|
Nguyen KT, Holloway MP, Altura RA. The CRM1 nuclear export protein in normal development and disease. INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND MOLECULAR BIOLOGY 2012; 3:137-151. [PMID: 22773955 PMCID: PMC3388738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 05/16/2012] [Indexed: 06/01/2023]
Abstract
CRM1 (Chromosomal Maintenance 1, also known as Exportin 1) is the major mammalian export protein that facilitates the transport of large macromolecules including RNA and protein across the nuclear membrane to the cytoplasm. The gene encoding CRM1 was originally identified in yeast as required to maintain higher order chromosome structure. In mammalian cells, CRM1 was found to bind several nuclear pore proteins hence its role in nuclear-cytosolic transport was discovered. In addition to nuclear-cytosolic transport, CRM1 also plays a role in centrosome duplication and spindle assembly, especially in response to DNA damage. The crystal structure of CRM1 suggests a complex protein that binds the Ran protein bound to GTP, allowing for a conformational change that facilitates binding to different cargo proteins through a nuclear export signal (NES). Included in the cadre of cargo are multiple tumor suppressor and oncoproteins as p53, BRCA1, Survivin, NPM, and APC, which function in the nucleus to regulate transcription or aid in chromosomal assembly and movement. An imbalance in the cytosolic level of these proteins has been observed in cancer cells, resulting in either inactivation (tumor suppressor) or an excess of anti-apoptotic activity (oncoprotein). Thus, the concept of inhibiting CRM1 has been explored as a potential therapeutic intervention. Indeed, inhibition of CRM1 by a variety of small molecules that interfere with cargo-NES binding results in cancer cell death. Whether all of these proteins together are responsible for this phenotype or whether specific proteins are required for this effect is unclear at this time.
Collapse
Affiliation(s)
- Kevin T Nguyen
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, Hasbro Children's Hospital and The Warren Alpert Medical School at Brown University Providence, Rhode Island, USA
| | | | | |
Collapse
|
28
|
Expression of Spy1 protein in human non-Hodgkin's lymphomas is correlated with phosphorylation of p27 Kip1 on Thr187 and cell proliferation. Med Oncol 2012; 29:3504-14. [PMID: 22492278 DOI: 10.1007/s12032-012-0224-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Accepted: 03/19/2012] [Indexed: 12/28/2022]
Abstract
Aberrations in cell cycle control are often observed in tumors and might even be necessary in tumor development. Spy1, a novel cell cycle regulatory protein, can control cell progression and survival through the atypical activation of cyclin-dependent kinases (CDKs). In this progression, the phosphorylation of p27(Kip1) at Thr187 by CDK2 was shown to be a chief role. In this study, we studied 183 human specimens including reactive lymphoid and Non-Hodgkin's Lymphomas (NHLs) tissues. Immunohistochemistry (IHC) analysis suggested that Spy1 and pThr187-p27 were overexpressed in NHLs. The expression of Spy1 was positively related to pThr187-p27 and proliferation marker Ki-67 expression. In a multivariate analysis, high Spy1 and pThr187-p27 expressions were showed to be associated with poor prognosis in NHLs. While in vitro, following release of Jurkat cells from serum starvation, the expression of Spy1 was upregulated, as well as pThr187-p27 and CDK2. And an increased interaction between Spy1 and pThr187-p27 was demonstrated at 4 h after serum stimulation. Additionally, transfecting cells with Spy1-siRNA could diminish the expression of pThr187-p27 and arrest cell growth. Our results suggest that Spy1 may be a possible prognostic indicator in NHLs, and it was correlated with phosphorylation of p27(Kip1) on Thr187. These findings provide a rational framework for further development of Spy1 inhibitors as a novel class of anti-tumor agents.
Collapse
|
29
|
Ueberham U, Hilbrich I, Ueberham E, Rohn S, Glöckner P, Dietrich K, Brückner MK, Arendt T. Transcriptional control of cell cycle-dependent kinase 4 by Smad proteins--implications for Alzheimer's disease. Neurobiol Aging 2012; 33:2827-40. [PMID: 22418736 DOI: 10.1016/j.neurobiolaging.2012.01.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 01/23/2012] [Accepted: 01/25/2012] [Indexed: 01/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by deregulation of neuronal cell cycle and differentiation control eventually resulting in cell death. During brain development, neuronal differentiation is regulated by Smad proteins, which are elements of the canonical transforming growth factor β (TGF-β) signaling pathway, linking receptor activation to gene expression. In the normal adult brain, Smad proteins are constitutively phosphorylated and predominantly localized in neuronal nuclei. Under neurodegenerative conditions such as AD, the subcellular localization of their phosphorylated forms is heavily disturbed, raising the question of whether a nuclear Smad deficiency in neurons might contribute to a loss of neuronal differentiation control and subsequent cell cycle re-entry. Here, we show by luciferase reporter assays, electromobility shift, and RNA interference (RNAi) technique a direct binding of Smad proteins to the CDK4 promoter inducing transcriptional inhibition of cell cycle-dependent kinase 4 (Cdk4). Mimicking the neuronal deficiency of Smad proteins observed in AD in cell culture by RNAi results in elevation of Cdk4 and retardation of neurite outgrowth. The results identify Smad proteins as direct transcriptional regulators of Cdk4 and add further evidence to a Smad-dependent deregulation of Cdk4 in AD, giving rise to neuronal dedifferentiation and cell death.
Collapse
Affiliation(s)
- Uwe Ueberham
- Department for Molecular and Cellular Mechanisms of Neurodegeneration, Universität Leipzig, Paul Flechsig Institute of Brain Research, Leipzig, D-04109, Jahnallee 59, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Sterlacci W, Fiegl M, Tzankov A. Prognostic and Predictive Value of Cell Cycle Deregulation in Non-Small-Cell Lung Cancer. Pathobiology 2012; 79:175-94. [DOI: 10.1159/000336462] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 01/12/2012] [Indexed: 12/29/2022] Open
|
31
|
Wang ML, Walsh R, Robinson KL, Burchard J, Bartz SR, Cleary M, Galloway DA, Grandori C. Gene expression signature of c-MYC-immortalized human fibroblasts reveals loss of growth inhibitory response to TGFβ. Cell Cycle 2011; 10:2540-8. [PMID: 21720214 DOI: 10.4161/cc.10.15.16309] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cancer cells exhibit the ability to proliferate indefinitely, but paradoxically, overexpression of cellular oncogenes in primary cells can result in a rapid and irreversible cell cycle arrest known as oncogene-induced senescence (OIS). However, we have shown that constitutive overexpression of the oncogene c-MYC in primary human foreskin fibroblasts results in a population of cells with unlimited lifespan; these immortalized cells are henceforth referred to as iMYC. Here, in order to further elucidate the mechanisms underlying the immortalization process, a gene expression signature of three independently established iMYC cell lines compared to matched early passage c-MYC overexpressing cells was derived. Network analysis of this "iMYC signature" indicated that a large fraction of the down-regulated genes were functionally connected and major nodes centered around the TGFβ, IL-6 and IGF-1 signaling pathways. Here, we focused on the functional validation of the alteration of TGFβ response during c-MYC-mediated immortalization. The results demonstrate loss of sensitivity of iMYC cells to activation of TGFβ signaling upon ligand addition. Furthermore, we show that aberrant regulation of the p27 tumor suppressor protein in iMYC cells is a key event that contributes to loss of response to TGFβ. These findings highlight the potential to reveal key pathways contributing to the self-renewal of cancer cells through functional mining of the unique gene expression signature of cells immortalized by c-MYC.
Collapse
Affiliation(s)
- Myra L Wang
- Program in Cancer Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Thenappan A, Shukla V, Khalek FJA, Li Y, Shetty K, Liu P, Li L, Johnson RL, Johnson L, Mishra L. Loss of transforming growth factor β adaptor protein β-2 spectrin leads to delayed liver regeneration in mice. Hepatology 2011; 53:1641-50. [PMID: 21520177 PMCID: PMC3162320 DOI: 10.1002/hep.24111] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
UNLABELLED Liver regeneration, following partial hepatectomy (PHx), occurs through precisely controlled and synchronized cell proliferation, in which quiescent hepatocytes undergo one to two rounds of replication, with restoration of liver mass and function. We previously demonstrated that loss of the Smad3/4 adaptor protein β-2 spectrin (β2SP) is associated with faster entry into S phase, and hepatocellular cancer formation. These observations led us to further pursue the role of β2SP in cell cycle progression in vivo. Liver regeneration studies with PHx in β2SP(+/-) mice reveal a surprising and significant decrease in liver/body weight ratio at 48 hours after PHx in β2SP(+/-) mice in comparison to wildtype mice. At 48 hours after PHx we also observe decreased levels of cyclin E (2.4-fold, P < 0.05), Cdk1 (7.2-fold, P < 0.05), cyclin A, pRb (Ser249/Thr252), proliferative cell nuclear antigen (PCNA), cyclin D1 with elevated levels of pCdk1 (Thr14) (3.6-fold, P < 0.05). Strikingly, at 24 hours elevated levels of p53 (4-fold, P < 0.05), phospho-p53 (ser15 and ser20), and p21 (200-fold, P < 0.05) persisting to 48 hours after PHx further correlated with raised expression of the DNA damage markers pChk2 (Thr68) and γH2AX (S139). However, compromised cell cycle progression with loss of β2SP is not rescued by inhibiting p53 function, and that G(2) /M phase arrest observed is independent and upstream of p53. CONCLUSION β2SP deficiency results in dysfunctional hepatocyte cell cycle progression and delayed liver regeneration at 48 hours after PHx, which is p53-independent. β2SP loss may increase susceptibility to DNA damage, impair cell cycle progression, and ultimately lead to hepatocellular cancer.
Collapse
Affiliation(s)
- Arun Thenappan
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Vivek Shukla
- Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Feras J Abdul Khalek
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Ying Li
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kirti Shetty
- Institute of Transplantation, Hepatobiliary Diseases and Surgery, Georgetown University Medical Center, Washington, DC
| | - Pu Liu
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lu Li
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Randy L Johnson
- Department of Biochemistry and Molecular Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lynt Johnson
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, Institute of Transplantation, Hepatobiliary Diseases and Surgery, Georgetown University Medical Center, Washington, DC
| | - Lopa Mishra
- Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas,Correspondence to: Lopa Mishra, Lopa Mishra, MD, Del & Dennis McCarthy Distinguished Professor and Chair, Department of Gastroenterology, Hepatology, and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1466, Houston, TX, 77030, Tel: 202-687-5707, Fax: 202-687-0992,
| |
Collapse
|
33
|
Borriello A, Bencivenga D, Criscuolo M, Caldarelli I, Cucciolla V, Tramontano A, Borgia A, Spina A, Oliva A, Naviglio S, Della Ragione F. Targeting p27Kip1 protein: its relevance in the therapy of human cancer. Expert Opin Ther Targets 2011; 15:677-93. [PMID: 21355788 DOI: 10.1517/14728222.2011.561318] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Cell division cycle progression is achieved by a sequential and stringently concerted activation of a family of serine-threonine kinases, namely the cyclin-dependent kinases (CDKs). p27(Kip1) is a pivotal CDK inhibitor and a tight modulator of CDK-dependent phenotypes. Thus, p27(Kip1) plays a fundamental role in key cellular processes such as proliferation, differentiation, apoptosis, substrate adhesion and motility. Intriguingly, when p27(Kip1) is localized in the nucleus, it acts as an antiproliferative protein, while, in the cytosol, p27(Kip1) promotes cytoskeleton remodeling and might positively influence metastatization. Downregulation of p27(Kip1) nuclear level or its cytosolic mislocalization are consistently correlated with poor prognosis of numerous types of human epithelial and non-epithelial cancers. AREAS COVERED This review illustrates the basic structural features of p27(Kip1) protein, its metabolism and alterations in human malignancies, along with describing anticancer strategies based on targeting p27(Kip1). EXPERT OPINION Given the role of p27(Kip1) in the control of cell proliferation and its decreased level observed in malignancies with poor outcome, drugs able to handle the protein levels and localization might represent an important goal for novel specific and effective anticancer strategies. Although no convincing proofs have been reported, putative negative consequences of p27(Kip1) targeting might be also conceivable.
Collapse
Affiliation(s)
- Adriana Borriello
- Second University of Naples, Medical School, Department of Biochemistry and Biophysics F. Cedrangolo, Via De Crecchio 7, 80138 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Recent progress in the field of molecular biology has allowed us to identify at least two different molecular mechanisms implicated in colorectal carcinogenesis (CRC): chromosomal instability (CIN) and genetic instability. Even though the two molecular mechanisms differ, their signalling pathways, implicated in malignant transformation of colonic epithelial cells, appear to be similar. The most frequent group of CRC, which represents 80% of sporadic CRC, is characterized by allelic losses on the short arm of chromosome 17 and 8 and on the long arm of chromosome 5, 18 and 22. These allelic losses are associated with mutations in TP53, APC, SMAD2 and SMAD4 genes. All of these alterations are grouped under the phenotype CIN. A genetic instability termed MSI (microsatellite instability), which results from a mismatch repair (MMR) deficiency, appears in 12-15% of CRC cases. The presence of MMR deficiency leads to the accumulation of mutations in genes controlling cell cycle and apoptosis (TGFBRII, BAX or CASPASE5). More recently, the existence of a third phenotype was suggested. The main alteration associated with this group of tumors is the hypermethylation of the promoter region of numerous genes, leading to their inactivation. An activating mutation of BRAF is frequently associated with this phenotype. As described above, CRC shows genetic heterogeneity, however the consequences in terms of signalling pathway alterations are similar. For example, the activation of Wnt signalling pathways can result from the inactivation of the APC gene in the CIN phenotype or from an activating mutation in the β-catenin gene in MSI tumors. The inactivation of TGFβ pathways is also present in both tumor types and is driven by SMAD4, and more rarely by a SMAD2 inactivating mutation in CIN tumors, or by the existence of a frame-shift mutation occurring in a polyG coding track of the TGFβ (transforming growth factor) receptor type II in MSI tumors. The RAS-MAP kinase pathway is activated by KRAS mutations in CIN tumors or by BRAF mutations in MSI tumors. The p53 pathway is inactivated by TP53 inactivation in CIN tumors or by BAX inactivating mutations in MSI tumors.
Collapse
|
35
|
Hershko DD. Cyclin-dependent kinase inhibitor p27 as a prognostic biomarker and potential cancer therapeutic target. Future Oncol 2010; 6:1837-47. [PMID: 21142858 DOI: 10.2217/fon.10.144] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The prognosis and clinical management of patients with cancer is commonly determined by traditional clinical and pathological factors. Nevertheless, patients may present with significantly different clinical outcomes despite similar clinicopathological features. This has prompted intense research to find biological markers that may closely reflect tumor biology and thereby clinical outcome. This article presents the current knowledge on the prognostic significance of p27 expression in cancer and its potential role as a target for future therapy.
Collapse
Affiliation(s)
- Dan D Hershko
- Department of Surgery & Breast Health Institute, Rambam Health Care Campus & the Technion – Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
36
|
Klopfleisch R, Schütze M, Gruber A. Loss of p27 expression in canine mammary tumors and their metastases. Res Vet Sci 2010; 88:300-3. [DOI: 10.1016/j.rvsc.2009.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 07/30/2009] [Accepted: 08/18/2009] [Indexed: 10/20/2022]
|
37
|
Abstract
Acute kidney injury (AKI) activates pathways of cell death and cell proliferation. Although seemingly discrete and unrelated mechanisms, these pathways can now be shown to be connected and even to be controlled by similar pathways. The dependence of the severity of renal-cell injury on cell cycle pathways can be used to control and perhaps to prevent acute kidney injury. This review is written to address the correlation between cellular life and death in kidney tubules, especially in acute kidney injury.
Collapse
|
38
|
Luca F, Kashyap S, Southard C, Zou M, Witonsky D, Di Rienzo A, Conzen SD. Adaptive variation regulates the expression of the human SGK1 gene in response to stress. PLoS Genet 2009; 5:e1000489. [PMID: 19461886 PMCID: PMC2679193 DOI: 10.1371/journal.pgen.1000489] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 04/22/2009] [Indexed: 12/22/2022] Open
Abstract
The Serum and Glucocorticoid-regulated Kinase1 (SGK1) gene is a target of the glucocorticoid receptor (GR) and is central to the stress response in many human tissues. Because environmental stress varies across habitats, we hypothesized that natural selection shaped the geographic distribution of genetic variants regulating the level of SGK1 expression following GR activation. By combining population genetics and molecular biology methods, we identified a variant (rs9493857) with marked allele frequency differences between populations of African and European ancestry and with a strong correlation between allele frequency and latitude in worldwide population samples. This SNP is located in a GR-binding region upstream of SGK1 that was identified using a GR ChIP-chip. SNP rs9493857 also lies within a predicted binding site for Oct1, a transcription factor known to cooperate with the GR in the transactivation of target genes. Using ChIP assays, we show that both GR and Oct1 bind to this region and that the ancestral allele at rs9493857 binds the GR-Oct1 complex more efficiently than the derived allele. Finally, using a reporter gene assay, we demonstrate that the ancestral allele is associated with increased glucocorticoid-dependent gene expression when compared to the derived allele. Our results suggest a novel paradigm in which hormonal responsiveness is modulated by sequence variation in the regulatory regions of nuclear receptor target genes. Identifying such functional variants may shed light on the mechanisms underlying inter-individual variation in response to environmental stressors and to hormonal therapy, as well as in the susceptibility to hormone-dependent diseases.
Collapse
Affiliation(s)
- Francesca Luca
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Sonal Kashyap
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - Catherine Southard
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Min Zou
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| | - David Witonsky
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Anna Di Rienzo
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Suzanne D. Conzen
- Department of Medicine, The University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
39
|
Bertagnolli MM, Warren RS, Niedzwiecki D, Mueller E, Compton CC, Redston M, Hall M, Hahn HP, Jewell SD, Mayer RJ, Goldberg RM, Saltz LB, Loda M. p27Kip1 in stage III colon cancer: implications for outcome following adjuvant chemotherapy in cancer and leukemia group B protocol 89803. Clin Cancer Res 2009; 15:2116-22. [PMID: 19276255 PMCID: PMC3059545 DOI: 10.1158/1078-0432.ccr-08-2674] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND In retrospective studies, loss of p27(Kip1) (p27), a cyclin-dependent kinase inhibitor, has been associated with poor prognosis following colorectal cancer treatment. In a prospective study, we validated this relationship in patients enrolled on a trial of adjuvant chemotherapy for stage III colon cancer. METHODS Cancer and Leukemia Group B protocol 89803 randomized 1,264 stage III colon cancer patients to receive weekly bolus 5-fluorouracil/leucovorin or weekly bolus irinotecan, 5-fluorouracil, and leucovorin (IFL). The primary endpoint was overall survival (OS); disease-free survival was a secondary endpoint. Expression of p27 and DNA mismatch repair proteins were determined by immunohistochemistry in primary tumor and normal tissue from paraffin blocks. Data were analyzed using log-rank test. RESULTS Of 601 tumors analyzed, 207 (34.4%) showed p27 loss, 377 (62.8%) retained p27, and 17 (2.8%) were indeterminate. Patients with p27-negative tumors showed reduced OS [5-year OS 66%: 95% confidence interval (95% CI), 0.59-0.72 versus 75%: 95% CI, 0.70-0.79; log-rank P = 0.021]. This relationship was not influenced by treatment arm. Combination of p27 status with mismatch repair status, however, identified a small subset of patients that may benefit from IFL (n = 36; 5-year disease-free survival 81%: 95% CI, 0.64-0.98 versus 47%: 95% CI, 0.21-0.72; log-rank P = 0.042; 5-year OS 81%: 95% CI, 0.64-0.98 versus 60%: 95% CI, 0.35-0.85; log-rank P = 0.128). CONCLUSIONS Loss of p27 is associated with reduced survival in stage III colon cancer but by itself does not indicate a significant difference in outcome between patients treated IFL or 5-fluorouracil/leucovorin.
Collapse
|
40
|
Hershko DD. Oncogenic properties and prognostic implications of the ubiquitin ligase Skp2 in cancer. Cancer 2008; 112:1415-24. [PMID: 18260093 DOI: 10.1002/cncr.23317] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The expression of Skp2, the ubiquitin ligase subunit that targets p27(Kip1) for degradation, is commonly overexpressed in human cancers. p27(Kip1) is a negative regulator of the cell cycle that plays an important role in tumor suppression. Loss of p27(Kip1) secondary to enhanced ubiquitin-mediated degradation results in uncontrolled proliferation and promotes tumor progression. In the present study the prognostic implications of Skp2 are reviewed and the mechanisms that regulate its expression in different human cancers. A review and analysis of the English literature was undertaken. Overexpression of Skp2 mRNA and protein levels was observed in many aggressive cancers and was commonly associated with down-regulation of p27(Kip1) levels and loss of tumor differentiation. Skp2 is an independent prognostic marker for disease-free and overall survival and may provide additional predictive information to that provided by p27(Kip1) alone. Targeting Skp2 in experimental models resulted in up-regulation of p27(Kip1) and arrested cellular proliferation. Alterations in Skp2 expression have profound effects on cancer progression and may serve as an accurate and independent prognostic marker. Thus, determination of levels of Skp2 and p27(Kip1) by readily available immunohistochemical studies may be a useful tool in the assessment of prognosis, especially in patients with intermediate disease, and may potentially assist in the planning of adjuvant therapy. Skp2 may be an attractive target for the development of novel interventional therapy.
Collapse
Affiliation(s)
- Dan D Hershko
- Department of Surgery and Breast Health Institute, Rambam Health Care Campus and the Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
41
|
Watkin H, Richert MM, Lewis A, Terrell K, McManaman JP, Anderson SM. Lactation failure in Src knockout mice is due to impaired secretory activation. BMC DEVELOPMENTAL BIOLOGY 2008; 8:6. [PMID: 18215306 PMCID: PMC2266720 DOI: 10.1186/1471-213x-8-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Accepted: 01/23/2008] [Indexed: 11/17/2022]
Abstract
Background Mammary gland development culminates in lactation and is orchestrated by numerous stimuli and signaling pathways. The Src family of nonreceptor tyrosine kinases plays a pivotal role in cell signaling. In order to determine if Src plays a role in mammary gland development we have examined mammary gland development and function during pregnancy and lactation in mice in which expression of Src has been eliminated. Results We have characterized a lactation defect in the Src-/- mice which results in the death of over 80% of the litters nursed by Src-/- dams. Mammary gland development during pregnancy appears normal in these mice; however secretory activation does not seem to occur. Serum prolactin levels are normal in Src-/- mice compared to wildtype controls. Expression of the prolactin receptor at both the RNA and protein level was decreased in Src-/- mice following the transition from pregnancy to lactation, as was phosphorylation of STAT5 and expression of milk protein genes. These results suggest that secretory activation, which occurs following parturition, does not occur completely in Src-/- mice. Failed secretory activation results in precocious involution in the mammary glands of Src-/- even when pups were suckling. Involution was accelerated following pup withdrawal perhaps as a result of incomplete secretory activation. In vitro differentiation of mammary epithelial cells from Src-/- mice resulted in diminished production of milk proteins compared to the amount of milk proteins produced by Src+/+ cells, indicating a direct role for Src in regulating the transcription/translation of milk protein genes in mammary epithelial cells. Conclusion Src is an essential signaling modulator in mammary gland development as Src-/- mice exhibit a block in secretory activation that results in lactation failure and precocious involution. Src appears to be required for increased expression of the prolactin receptor and successful downstream signaling, and alveolar cell organization.
Collapse
Affiliation(s)
- Harriet Watkin
- Department of Pathology, University of Colorado Health Sciences Center, Research Complex I, South Tower, Mail Stop 8104, 12801 East 17th Avenue, Aurora, CO 80045, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Nozoe T, Oyama T, Takenoyama M, Hanagiri T, Sugio K, Yasumoto K. Significance of Immunohistochemical Expression of p27 and Involucrin as the Marker of Cellular Differentiation of Squamous Cell Carcinoma of the Esophagus. Oncology 2007; 71:402-10. [PMID: 17878746 DOI: 10.1159/000108611] [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] [Received: 01/30/2007] [Accepted: 06/21/2007] [Indexed: 01/18/2023]
Abstract
PURPOSE p27kip1 belongs to the KIP/CIP family of cyclin-dependent kinase inhibitors and is considered to be a tumor suppressor. Involucrin has been known as a marker of differentiation of squamous cell carcinoma (SCC). The aim of this study was to evaluate the clinicopathologic significance of the expression of p27 and involucrin in esophageal SCC. METHODS Immunohistochemical expression of p27 and involucrin was examined in 70 specimens of esophageal SCC. The correlation of the expression of these proteins and clinicopathologic features was evaluated. RESULTS Cellular differentiation in esophageal SCC was significantly correlated with the expression of p27 and involucrin (p = 0.010 and p = 0.002, respectively). Among well, moderately and poorly differentiated SCCs, 45.8 +/- 21.6, 20.0 +/- 15.0 and 10.6 +/- 9.1% of carcinoma cells expressed involucrin, respectively (p < 0.0001 for well vs. poorly, p < 0.0001 for well vs. moderately, and p = 0.042 for moderately vs. poorly). There existed a more powerful statistical difference regarding the histological grade between SCCs with the expression of both p27 and involucrin and tumors with other expression patterns (p = 0.0001). CONCLUSIONS Expression of both p27 and involucrin can be a powerful biological marker of cellular differentiation of esophageal SCC.
Collapse
Affiliation(s)
- Tadahiro Nozoe
- Second Department of Surgery, University of Occupational and Environmental Health, Kitakyushu, Japan.
| | | | | | | | | | | |
Collapse
|
43
|
Auld CA, Fernandes KM, Morrison RF. Skp2-mediated p27(Kip1) degradation during S/G2 phase progression of adipocyte hyperplasia. J Cell Physiol 2007; 211:101-11. [PMID: 17096381 DOI: 10.1002/jcp.20915] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
p27(Kip1), an important regulator of Cdk2 activity and G1/S transition, is tightly regulated in a cell-type and condition-specific manner to integrate mitogenic and differentiation signals governing cell cycle progression. We show that p27 protein levels progressively declined from mid-G1 through late-G2 phase as density-arrested 3T3-L1 preadipocytes synchronously reentered the cell cycle during early stages of adipocyte differentiation. This dramatic fall in p27 protein accumulation was due, at least in part, to a decrease in protein stability. Specific inhibitors of the 26S proteasome were shown to completely block the decrease in p27 protein levels throughout G1, increase the abundance of ubiquitylated p27 protein, and inhibit G1/S transition resulting in G1 arrest. It is further demonstrated that p27 was phosphorylated on threonine 187 during S phase progression by Cdk2 and that phosphorylated p27 was polyubiquitylated and degraded. Furthermore, we demonstrate that Skp2 and Cks1 dramatically increased during S/G2 phase progression concomitantly with the maximal fall in p27 protein. Complete knockdown of Skp2 with RNA interference partially prevented p27 degradation equivalent to that observed with Cdk2 blockade suggesting that the SCF(Skp2) E3 ligase and other proteasome-dependent mechanisms contribute to p27 degradation during preadipocyte replication. Interestingly, Skp2-mediated p27 degradation was not essential for G1/S or S/G2 transition as preadipocytes shifted from quiescence to proliferation during adipocyte hyperplasia. Finally, evidence is presented suggesting that elevated p27 protein in the absence of Skp2 was neutralized by sequestration of p27 protein into Cyclin D1/Cdk4 complexes.
Collapse
Affiliation(s)
- Corinth A Auld
- Department of Nutrition, The University of North Carolina at Greensboro, Greensboro, North Carolina 27402, USA
| | | | | |
Collapse
|
44
|
Huang X, Zhu D, Lou Y. A novel anticancer agent, icaritin, induced cell growth inhibition, G1 arrest and mitochondrial transmembrane potential drop in human prostate carcinoma PC-3 cells. Eur J Pharmacol 2007; 564:26-36. [PMID: 17382317 DOI: 10.1016/j.ejphar.2007.02.039] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 01/26/2007] [Accepted: 02/01/2007] [Indexed: 01/29/2023]
Abstract
Icariin and icaritin with prenyl group have been demonstrated for their selective estrogen receptor modulating activities. We screened their effects on cell growth in human prostate carcinoma PC-3 cell line (estrogen receptor positive) in vitro. PC-3 cell line was used for the measurement of anti-carcinoma activities of 0-100 micromol/l icaritin and 30 micromol/l icariin. 1 micromol/l 17-beta estradiol (E(2)) served as the estrogen positive control, and 1 micromol/l ICI 182,780 [7 alpha-[9 (4,4,5,5,5-pentafluoropentyl) sulfinyl] nonyl]-estra-1,3,5(10)-triene-3,17h-diol]] served as the specific estrogen receptor antagonist. Primary cultured rat prostate basal cells used as cell growth selective control. The growth-inhibitory effects were analyzed using MTT assay, and fluorochrome staining, flow cytometry, and immunoblotting were employed to illustrate the possible mechanisms. When treated with icaritin for 24 to 72 h, cell growth was strongly inhibited (at 48 h IC(50) was 10.74+/-1.59 micromol/l, P<0.001) companied with a mitochondrial transmembrane potential (_Psim) drop. Meanwhile, few changes in IC(50) could be observed when co-incubated with ICI 182,780. Icaritin-induced growth inhibition was associated with G(1) arrest (P<0.05), and G(2)-M arrest depending upon doses. Consistently with G(1) arrest, icaritin increased protein expressions of pRb, p27(Kip1) and p16(Ink4a), while showed decrease in phosphorylated pRb, Cyclin D1 and CDK4. Comparatively, icariin has much lower effects on PC-3 cells and showed only weak G(1) arrest, suggesting a possible structure-activity relationship. These findings suggested a novel anticancer efficacy of icaritin mediated selectively via induction of cell cycle arrest but not associated with estrogen receptors in PC-3 cells.
Collapse
Affiliation(s)
- Xin Huang
- Institute of Pharmacology & Toxicology and Biochemical Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 353 Yan'an Road, Hangzhou 310031, China.
| | | | | |
Collapse
|
45
|
Chu I, Sun J, Arnaout A, Kahn H, Hanna W, Narod S, Sun P, Tan CK, Hengst L, Slingerland J. p27 phosphorylation by Src regulates inhibition of cyclin E-Cdk2. Cell 2007; 128:281-94. [PMID: 17254967 PMCID: PMC1961623 DOI: 10.1016/j.cell.2006.11.049] [Citation(s) in RCA: 288] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 10/03/2006] [Accepted: 11/20/2006] [Indexed: 01/02/2023]
Abstract
The kinase inhibitor p27Kip1 regulates the G1 cell cycle phase. Here, we present data indicating that the oncogenic kinase Src regulates p27 stability through phosphorylation of p27 at tyrosine 74 and tyrosine 88. Src inhibitors increase cellular p27 stability, and Src overexpression accelerates p27 proteolysis. Src-phosphorylated p27 is shown to inhibit cyclin E-Cdk2 poorly in vitro, and Src transfection reduces p27-cyclin E-Cdk2 complexes. Our data indicate that phosphorylation by Src impairs the Cdk2 inhibitory action of p27 and reduces its steady-state binding to cyclin E-Cdk2 to facilitate cyclin E-Cdk2-dependent p27 proteolysis. Furthermore, we find that Src-activated breast cancer lines show reduced p27 and observe a correlation between Src activation and reduced nuclear p27 in 482 primary human breast cancers. Importantly, we report that in tamoxifen-resistant breast cancer cell lines, Src inhibition can increase p27 levels and restore tamoxifen sensitivity. These data provide a new rationale for Src inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Isabel Chu
- Braman Family Breast Cancer Institute, and Department of Biochemistry and Molecular Biology, U. of Miami Miller School of Medicine, Miami, Florida; U.S.A
- Departments of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jun Sun
- Braman Family Breast Cancer Institute, and Department of Biochemistry and Molecular Biology, U. of Miami Miller School of Medicine, Miami, Florida; U.S.A
| | - Angel Arnaout
- Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Harriette Kahn
- Pathobiology and Lab Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Wedad Hanna
- Pathobiology and Lab Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Steven Narod
- Pathobiology and Lab Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ping Sun
- Pathobiology and Lab Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Cheng-Keat Tan
- Braman Family Breast Cancer Institute, and Department of Biochemistry and Molecular Biology, U. of Miami Miller School of Medicine, Miami, Florida; U.S.A
| | - Ludger Hengst
- Division of Medical Biochemistry, Biocenter - Innsbruck Medical University, Innsbruck, Austria
| | - Joyce Slingerland
- Braman Family Breast Cancer Institute, and Department of Biochemistry and Molecular Biology, U. of Miami Miller School of Medicine, Miami, Florida; U.S.A
- Departments of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Gabellini C, Pucci B, Valdivieso P, D'Andrilli G, Tafani M, De Luca A, Masciullo V. p27kip1 overexpression promotes paclitaxel-induced apoptosis in pRb-defective SaOs-2 cells. J Cell Biochem 2006; 98:1645-52. [PMID: 16598766 DOI: 10.1002/jcb.20900] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
p27kip1 is a cyclin-dependent kinase (CDK) inhibitor, which controls several cellular processes in strict collaboration with pRb. We evaluated the role of p27kip1 in paclitaxel-induced apoptosis in the pRb-defective SaOs-2 cells. Following 48 h of exposure of SaOs-2 cells to 100 nM paclitaxel, we observed an increase in p27kip1 expression caused by the decrease of the ubiquitin-proteasome activity. Such increase was not observed in SaOs-2 cells treated with the caspase inhibitors Z-VAD-FMK, suggesting that p27kip1 enhancement at 48 h is strictly related to apoptosis. Finally, we demonstrated that SaOs-2 cells transiently overexpressing the p27kip1 protein are more susceptible to paclitaxel-induced apoptosis than SaOs-2 cells transiently transfected with the empty vector. Indeed, after 48 h of paclitaxel treatment, 41.8% of SaOs-2 cells transiently transfected with a pcDNA3-p27kip1 construct were Annexin V-positive compared to 30.6% of SaOs-2 cells transfected with the empty vector (P < 0.05). In conclusion, we demonstrated that transfection of the pRb-defective SaOs-2 cells with the p27kip1 gene via plasmid increases their susceptibility to paclitaxel-induced apoptosis. The promoting effect of p27kip1 overexpression on apoptosis makes p27kip1 and proteasomal inhibitors interesting tools for therapy in patients with pRb-defective cancers.
Collapse
Affiliation(s)
- Chiara Gabellini
- Experimental Chemotherapy Laboratory, Regina Elena Cancer Institute, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
47
|
Auld CA, Morrison RF. Evidence for cytosolic p27(Kip1) ubiquitylation and degradation during adipocyte hyperplasia. Obesity (Silver Spring) 2006; 14:2136-44. [PMID: 17189539 DOI: 10.1038/oby.2006.250] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Subcellular localization has been shown to play an important role in determining activity and accumulation of p27 protein during cell cycle progression. The purpose of this study was to examine p27 localization and ubiquitylation in relation to E3 ligase expression during adipocyte hyperplasia. RESEARCH METHODS AND PROCEDURES This study used the murine 3T3-L1 preadipocyte model to examine p27 regulation during synchronous cell cycle progression. Cell lysates were isolated over time after hormonal stimulation, fractionated to cytosolic and nuclear compartments, and immunoblotted for relative protein determinations. RESULTS Data presented in this study show that p27 was present in the cytosol and nucleus in density-arrested preadipocytes and that abundance in both compartments decreased in a phase-specific manner as preadipocytes synchronously re-entered the cell cycle during early phases of adipocyte differentiation. Blocking CRM1-mediated nuclear export did not prevent degradation, nor did it cause nuclear accumulation of p27, suggesting that distinct mechanisms mediating cytosolic and nuclear p27 degradation were involved. Treating preadipocytes with a potent and specific proteasome inhibitor during hormonal stimulation prevented Skp2 accumulation and p27(187) phosphorylation, which are essential events for SCF(Skp2) E3 ligase activity and nuclear p27 ubiquitylation during S/G(2) phase progression. Proteasome blockade also resulted in the first evidence of cytosolic p27 ubiquitylation during late G(1) phase as preadipocytes undergo the transition from quiescence to proliferation. DISCUSSION These data are consistent with the postulate that p27 is ubiquitylated and targeted for degradation by the 26S proteasome in a phase-specific manner by distinct ubiquitin E3 ligases localized to the cytosol and nucleus during adipocyte hyperplasia.
Collapse
Affiliation(s)
- Corinth A Auld
- Department of Nutrition, 318 Stone Building, UNC Greensboro, Greensboro, NC 27402, USA
| | | |
Collapse
|
48
|
Hershko DD, Shapira M. Prognostic role of p27Kip1 deregulation in colorectal cancer. Cancer 2006; 107:668-75. [PMID: 16826582 DOI: 10.1002/cncr.22073] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
p27Kip1, an inhibitor of cyclin-dependent kinases, is a negative cell cycle regulator that plays an important role in tumor suppression. Deregulation of p27 is commonly observed in many human cancers secondary to enhanced ubiquitin-mediated degradation, mediated and rate-limited by its specific ubiquitin ligase subunits Skp2 and Cks1. In the present study the prognostic implications of p27 and the mechanisms that down-regulate its expression in colorectal cancer (CRC) are reviewed. A review and analysis of the English literature was conducted. Loss of p27 was strongly associated with aggressive tumor behavior and poor clinical outcome in CRC. Overexpression of Skp2 and Cks1 was observed in aggressive CRC and is responsible for down-regulation of p27 levels. Both Skp2 and Cks1 were found to be independent prognostic markers for survival and provide predictive information additional to that provided by p27 alone. Deregulation of p27 has a profound effect on tumor progression in CRC and was found to be an accurate and independent prognostic marker. Thus, determination of levels of p27 and of its ubiquitin ligase subunits by readily available immunohistochemical studies may be a useful tool in the assessment of prognosis, especially in patients with intermediate disease, and may potentially assist in the planning of adjuvant therapy and development of novel interventional therapy.
Collapse
Affiliation(s)
- Dan D Hershko
- Department of Surgery, Rambam Medical Center, Haifa, Israel.
| | | |
Collapse
|
49
|
Gonzalez-Angulo AM, Guarneri V, Gong Y, Cristofanilli M, Morales-Vasquez F, Sneige N, Hortobagyi GN, Esteva FJ. Downregulation of the Cyclin-Dependent Kinase Inhibitor p27kip1 Might Correlate with Poor Disease-Free and Overall Survival in Inflammatory Breast Cancer. Clin Breast Cancer 2006; 7:326-30. [PMID: 17092400 DOI: 10.3816/cbc.2006.n.045] [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] [Indexed: 11/20/2022]
Abstract
PURPOSE The objective of this study was to evaluate whether p27kip1 downregulation is a prognostic factor in patients with inflammatory breast carcinoma (IBC). PATIENTS AND METHODS Fifty-eight patients with IBC were treated between January 1994 and July 2002. Median age was 49 years. Thirty-eight patients had baseline biopsy specimens. Patients received preoperative chemotherapy with FAC (5-fluorouracil/doxorubicin/cyclophosphamide; 34%) or FAC followed by a taxane (66%). All patients underwent mastectomies. All patients received radiation therapy and hormonal treatment when indicated. Expression level of p27kip1 was evaluated by indirect immunoperoxidase procedure. The p27kip1 was considered downregulated if nuclear staining was present in < 50% of the neoplastic cells. RESULTS Thirty-two patients (84%) had p27kip1-downregulated tumors, and 6 patients (17%) had p27kip1-normal tumors. Six patients (16%) exhibited a pathologic complete response. At a median follow-up of 43 months, 25 recurrences (66%) and 27 deaths (71%) occurred. Patients with p27kip1-downregulated tumors had fewer pathologic complete responses (9% vs. 50%; P = 0.03) and had lower 4-year recurrence-free survival (23% vs. 83%; P = 0.03) and overall survival rates (36% vs. 83%; P = 0.01). CONCLUSION The p27kip1 deregulation manifested by low protein cellular concentration might represent an adverse prognostic marker in IBC and could provide a valuable tool for selecting treatment for this aggressive disease.
Collapse
Affiliation(s)
- Ana Maria Gonzalez-Angulo
- Department of Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77230-1439, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Kim SS, Shetty K, Katuri V, Kitisin K, Baek HJ, Tang Y, Marshall B, Johnson L, Mishra B, Mishra L. TGF-beta signaling pathway inactivation and cell cycle deregulation in the development of gastric cancer: role of the beta-spectrin, ELF. Biochem Biophys Res Commun 2006; 344:1216-23. [PMID: 16650383 PMCID: PMC4211257 DOI: 10.1016/j.bbrc.2006.03.236] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2006] [Accepted: 03/22/2006] [Indexed: 01/10/2023]
Abstract
We have shown that loss of ELF, a stem cell adaptor protein, disrupts TGF-beta signaling through Smad3 and Smad4 localization. Notably elf(+/-)/smad4(+/-) mice develop gastric cancer presenting this as an important model for analyzing molecular event in gastric carcinogenesis. To gain further insight into the functional role of ELF in gastric cancer suppression, we carried out a detailed characterization of cell cycle events leading to gastric tumorigenesis. elf(-/-) cells and elf(+/-)/smad4(+/-) mice demonstrate a marked alteration of cell cycle regulators, such as Cdk4, K-Ras, and p21. Levels of Cdk4 increased compared to normal controls, suggesting loss of ELF results in functional abnormalities in cell cycle regulation. We further demonstrate that the elf(-/-) MEFs show a disruption of G1/S cell cycle transition and a significant reduction in senescence. Thus, in response to ELF deficiency, the abnormalities of G1/S checkpoint and senescence contribute their increment of susceptibility to malignant transformation.
Collapse
Affiliation(s)
- Sang Soo Kim
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | - Kirti Shetty
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | - Varalakshmi Katuri
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | - Krit Kitisin
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | - Hye Jung Baek
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | - Yi Tang
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | - Blair Marshall
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | - Lynt Johnson
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | - Bibhuti Mishra
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
| | - Lopa Mishra
- Laboratory of Cancer Genetics, Digestive Diseases, and Developmental Molecular Biology, Department of Surgery, Medicine, Lombardi Cancer Center, Georgetown University, Washington, DC, USA
- Department of Veterans Affairs, Washington, DC, USA
| |
Collapse
|