1
|
Drymiotou S, Theodorou E, Rallis KS, Nicolaides M, Sideris M. Molecular Biomarkers in Borderline Ovarian Tumors: Towards Personalized Treatment and Prognostic Assessment. Cancers (Basel) 2025; 17:545. [PMID: 39941911 PMCID: PMC11816664 DOI: 10.3390/cancers17030545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Borderline Ovarian Tumours (BOTs) are a heterogenous group of ovarian neoplasms which have increased mitotic activity but lack stromal invasion. We performed a narrative review of the literature, aiming to identify prognostic molecular biomarkers that can potentially be used for treatment personalisation. We identified and discussed BRAF/KRAS, Cancer Antigen 125 (Ca 125), Calprotectin, p16ink4a, and Microsatellite instability (MSI) as the most studied biomarkers related to BOTs. Overall, BRAF and KRAS mutations are associated with earlier-stage and favourable prognosis; KRASmt may indicate extraovarian disease in serous BOT (sBOT). Ca125, the only currently clinically used biomarker, can be assessed pre-operatively and has an established role in post-operative surveillance, especially when it is raised pre-operatively or a high potential for malignant transformation is suspected post-operatively. p16ink4a expression trends could also indicate the malignant transformation of the tumour. Calprotectin has an inferior specificity to Ca125 and is not yet established as a biomarker, whilst there is very limited evidence available for MSI. As new evidence is coming along with artificial intelligence platforms, these biomarkers can be integrated and used towards the development of a precision model for treatment stratification and counselling in women diagnosed with BOTs.
Collapse
Affiliation(s)
- Stefania Drymiotou
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK; (S.D.); (E.T.); (K.S.R.)
| | - Efthymia Theodorou
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK; (S.D.); (E.T.); (K.S.R.)
| | - Kathrine Sofia Rallis
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK; (S.D.); (E.T.); (K.S.R.)
| | - Marios Nicolaides
- Guy’s and St Thomas’ NHS Foundation Trust, Westminster Bridge Road, London SE1 7EH, UK;
| | - Michail Sideris
- Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AD, UK; (S.D.); (E.T.); (K.S.R.)
- Wolfson Institute of Population Health, Queen Mary University of London, Charterhouse Square Campus, Barbican, London EC1M 6BQ, UK
| |
Collapse
|
2
|
Rocha MA, Cardoso AL, Martins C, Mello MLS. Sodium valproate affects the expression of p16 INK4a and p21 WAFI/Cip1 cyclin‑dependent kinase inhibitors in HeLa cells. Oncol Lett 2024; 28:432. [PMID: 39049983 PMCID: PMC11268092 DOI: 10.3892/ol.2024.14563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024] Open
Abstract
p16INK4a and p21WAF1/Cip1 are cyclin-dependent kinase inhibitors involved in cell cycle control, which can function as oncogenes or tumor suppressors, depending on the context of various extracellular and intracellular signals, and cell type. In human papillomavirus-induced cervical cancer, p16 INK4a shows oncogenic activity and functions as a diagnostic marker of cervical neoplasia, whereas p21 WAF1/Cip1 acts as a tumor suppressor and its downregulation is associated with the progression of malignant transformation. Several histone deacetylase (HDAC) inhibitors promote the positive and negative regulation of a number of genes, including p16 INK4a and p21 WAF1/Cip1; however, the effects of sodium valproate (VPA) on these genes and on the proteins they encode remain uncertain in HeLa cervical cancer cells. In the present study, these effects were investigated in HeLa cells treated with 0.5 or 2 mM VPA for 24 h, using reverse transcription-quantitative PCR, confocal microscopy and western blotting. The results revealed a decrease in the mRNA expression levels of p16 INK4a and a tendency for p16INK4a protein abundance to decrease in the presence of 2 mM VPA. By contrast, an increase in the protein expression levels of p21WAF1/Cip1 was detected in the presence of 0.5 and 2 mM VPA. Furthermore, VPA was confirmed to inhibit HDAC activity and induce global hyperacetylation of histone H3. Notably, VPA was shown to suppress p16 INK4a, a biomarker gene of cervical carcinoma, and to increase the abundance of the tumor suppressor protein p21WAF1/Cip1, thus contributing to the basic knowledge regarding the antitumorigenic potential of VPA. Exploration of epigenetic changes associated with the promoters of p16 INK4a and p21 WAF1/Cip1, such as histone H3 methylation, may provide further information and improve the understanding of these findings.
Collapse
Affiliation(s)
- Marina Amorim Rocha
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, São Paulo 13083-862, Brazil
| | - Adauto Lima Cardoso
- Department of Structural and Functional Biology, Institute of Biosciences at Botucatu, São Paulo State University, Botucatu, São Paulo 18618-689, Brazil
| | - Cesar Martins
- Department of Structural and Functional Biology, Institute of Biosciences at Botucatu, São Paulo State University, Botucatu, São Paulo 18618-689, Brazil
| | - Maria Luiza S. Mello
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, São Paulo 13083-862, Brazil
| |
Collapse
|
3
|
Zhang B, Liu P, Sheng H, Guo Y, Han Y, Suo L, Yuan Q. New Insight into the Potential Protective Function of Sulforaphene against ROS-Mediated Oxidative Stress Damage In Vitro and In Vivo. Int J Mol Sci 2023; 24:13129. [PMID: 37685936 PMCID: PMC10487408 DOI: 10.3390/ijms241713129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Sulforaphene (SFE) is a kind of isothiocyanate isolated from radish seeds that can prevent free-radical-induced diseases. In this study, we investigated the protective effect of SFE on oxidative-stress-induced damage and its molecular mechanism in vitro and in vivo. The results of cell experiments show that SFE can alleviate D-gal-induced cytotoxicity, promote cell cycle transformation by inhibiting the production of reactive oxygen species (ROS) and cell apoptosis, and show a protective effect on cells with H2O2-induced oxidative damage. Furthermore, the results of mice experiments show that SFE can alleviate D-galactose-induced kidney damage by inhibiting ROS, malondialdehyde (MDA), and 4-hydroxyalkenals (4-HNE) production; protect the kidney against oxidative stress-induced damage by increasing antioxidant enzyme activity and upregulating the Nrf2 signaling pathway; and inhibit the activity of pro-inflammatory factors by downregulating the expression of Toll-like receptor 4 (TLR4)-mediated inflammatory response. In conclusion, this research shows that SFE has antioxidant effects, providing a new perspective for studying the anti-aging properties of natural compounds.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; (B.Z.); (P.L.); (H.S.); (Y.G.); (Y.H.); (L.S.)
| |
Collapse
|
4
|
Bai F, Liu X, Zhang X, Mao Z, Wen H, Ma J, Pei XH. p18INK4C and BRCA1 inhibit follicular cell proliferation and dedifferentiation in thyroid cancer. Cell Cycle 2023; 22:1637-1653. [PMID: 37345432 PMCID: PMC10361144 DOI: 10.1080/15384101.2023.2225938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 04/13/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023] Open
Abstract
Only 3% of thyroid cancers are medullary thyroid carcinomas (MTCs), the rest are follicular epithelial cell derived non-MTCs (NMTCs). A dysfunctional INK4-CDK4-RB pathway is detected in most of NMTCs. DNA repair defects and genome instability are associated with NMTC dedifferentiation and aggressiveness. Whether inactivation of the INK4-CDK4-RB pathway induces NMTCs and how differentiation of NMTC cells is controlled remain elusive. In this study, we generated p18Ink4c and Brca1 singly and doubly deficient mice as well as p16Ink4a and Brca1 singly and doubly deficient mice. By using these mice and human thyroid carcinoma cell lines, we discovered that loss of p18Ink4c, not p16Ink4a, in mice stimulated follicular cell proliferation and induced NMTCs. Depletion of Brca1 alone or both p16Ink4a and Brca1 did not induce thyroid tumor. Depletion of Brca1 in p18Ink4c null mice results in poorly differentiated and aggressive NMTCs with epithelial-mesenchymal transition (EMT) features and enhanced DNA damage. Knockdown of BRCA1 in thyroid carcinoma cells activated EMT and promoted tumorigenesis whereas overexpression of BRCA1 inhibited EMT. BRCA1 and EMT marker expression were inversely related in human thyroid cancers. Our finding, for the first time, demonstrates that inactivation of INK4-CDK4-RB pathway induces NMTCs and that Brca1 deficiency promotes dedifferentiation of NMTC cells. These results suggest that BRCA1 and p18INK4C collaboratively suppress thyroid tumorigenesis and progression and CDK4 inhibitors will be effective for treatment of INK4-inactivated or cyclin D-overexpressed thyroid carcinomas.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, China
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| | - Xu Zhang
- Department of Pathology, School of Basic Medicine, Lanzhou University, Lanzhou, China
| | - Zhuo Mao
- Department of Physiology, Shenzhen University Health Science Center, Shenzhen, China
| | - He Wen
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Jinshan Ma
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
- Department of Thoracic Surgery, Xinjiang Uigur Autonomous Region People’s Hospital, Xinjiang, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, the First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, China
- Dewitt Daughtry Family Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
5
|
INK4 cyclin-dependent kinase inhibitors as potential prognostic biomarkers and therapeutic targets in hepatocellular carcinoma. Biosci Rep 2022; 42:231524. [PMID: 35771229 PMCID: PMC9284345 DOI: 10.1042/bsr20221082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
The INK4 family is an important family of cyclin-dependent kinase inhibitors (CDKIs) and consists of CDKN2A, CDKN2B, CDKN2, and CDKN2D. Abnormal expression of CDKN2A has been reported in hepatocellular carcinoma (HCC) and is associated with the prognosis of patients and infiltration of immune cells. However, there is a lack of systematic research on the roles of the other INK4 family members in the diagnosis, prognosis, and immune regulation of HCC. Using online public databases and clinical samples, we comprehensively analyzed the INK4 family in HCC. All four INK4 proteins were overexpressed in HCC and correlated with advanced cancer stage and poor prognosis. INK4 expression accurately distinguished tumor from normal tissue, particularly CDKN2A and CDKN2C. The INK4 family participated in cell-cycle regulation and the DNA damage repair pathway, which inhibited genotoxic-induced apoptosis in tumorigenesis. INK4 proteins were positively correlated with the infiltration of immune cells (B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells) and immune checkpoints (CTLA-4, PD1, and PD-L1). CDKN2D had the highest correlation (correlation coefficient >0.3) with all the above-mentioned infiltrating immune cells and immune checkpoints, indicating that it may be useful as an immunotherapy target. The INK4 family was valuable for diagnosis and predicting the prognosis of HCC and participated in the occurrence, progression, and immune regulation of HCC, demonstrating its potential as a diagnostic and prognostic biomarker and therapeutic target in HCC.
Collapse
|
6
|
Pan M, Sun Q, Li C, Tai R, Shi X, Sun C. HOXA5 inhibits adipocytes proliferation through transcriptional regulation of Ccne1 and blocking JAK2/STAT3 signaling pathway in mice. Biochem Cell Biol 2022; 100:325-337. [PMID: 35623098 DOI: 10.1139/bcb-2021-0558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The highly regulated proliferation of adipocytes plays a momentous role in fat development and obesity. Hoxa5 is an important member of Hox family, its encoded protein is an important transcription factor related to development. And its differential expression in different adipose tissues seems to indicate that Hoxa5 may be involved in the regulation of adipocyte proliferation. In order to evaluate the regulation mechanism of Hoxa5 on adipocyte proliferation, we constructed a variety of Hoxa5 expression vectors in vivo and in vitro to explore its mechanism on adipocyte proliferation and its potential impact on obesity. We have observed that the overexpression of Hoxa5 strongly reduces cell counts, and Hoxa5 can inhibit cell proliferation and block cell cycle progression by regulating the expression of genes such as Cyclin E, Cycling D1 and p53. Most importantly, we demonstrated that Hoxa5 exerts its effect by regulating the signaling pathway of Janus kinase 2 (JAK2) signal transduction and transcription 3 (STAT3) activator, as well as binding to the promoter region of Ccne1 and inhibiting the transcription of Ccne1.This study provides an in-depth understanding of the potential molecular mechanism of Hoxa5 inhibiting adipocyte proliferation. Our results suggest the importance of Hoxa5 in the treatment of obesity.
Collapse
Affiliation(s)
- Miao Pan
- Northwest A&F University, 12469, Yangling, Shaanxi, China;
| | - Qian Sun
- Northwest A&F University, 12469, Yangling, Shaanxi, China;
| | - Chaowei Li
- Northwest A&F University, 12469, Yangling, Shaanxi, China;
| | - Ruiqing Tai
- Northwest A&F University, 12469, Yangling, Shaanxi, China;
| | - Xin'e Shi
- Northwest A&F University, 12469, Yangling, Shaanxi, China;
| | - Chao Sun
- Northwest A&F University, 12469, Yangling, Shaanxi, China;
| |
Collapse
|
7
|
Bai F, Zheng C, Liu X, Chan HL, Liu S, Ma J, Ren S, Zhu WG, Pei XH. Loss of function of GATA3 induces basal-like mammary tumors. Am J Cancer Res 2022; 12:720-733. [PMID: 34976209 PMCID: PMC8692904 DOI: 10.7150/thno.65796] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/13/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose: GATA3 is a transcription factor essential for mammary luminal epithelial cell differentiation. Expression of GATA3 is absent or significantly reduced in basal-like breast cancers. Gata3 loss-of-function impairs cell proliferation, making it difficult to investigate the role of GATA3 deficiency in vivo. We previously demonstrated that CDK inhibitor p18INK4c (p18) is a downstream target of GATA3 and restrains mammary epithelial cell proliferation and tumorigenesis. Whether and how loss-of-function of GATA3 results in basal-like breast cancers remains elusive. Methods: We generated mutant mouse strains with heterozygous germline deletion of Gata3 in p18 deficient backgrounds and developed a Gata3 depleted mammary tumor model system to determine the role of Gata3 loss in controlling cell proliferation and aberrant differentiation in mammary tumor development and progression. Results: Haploid loss of Gata3 reduced mammary epithelial cell proliferation with induction of p18, impaired luminal differentiation, and promoted basal differentiation in mammary glands. p18 deficiency induced luminal type mammary tumors and rescued the proliferative defect caused by haploid loss of Gata3. Haploid loss of Gata3 accelerated p18 deficient mammary tumor development and changed the properties of these tumors, resulting in their malignant and luminal-to-basal transformation. Expression of Gata3 negatively correlated with basal differentiation markers in MMTV-PyMT mammary tumor cells. Depletion of Gata3 in luminal tumor cells also reduced cell proliferation with induction of p18 and promoted basal differentiation. We confirmed that expression of GATA3 and basal markers are inversely correlated in human basal-like breast cancers. Conclusions: This study provides the first genetic evidence demonstrating that loss-of-function of GATA3 directly induces basal-like breast cancer. Our finding suggests that basal-like breast cancer may also originate from luminal type cancer.
Collapse
|
8
|
Bai F, Zhang LH, Liu X, Wang C, Zheng C, Sun J, Li M, Zhu WG, Pei XH. GATA3 functions downstream of BRCA1 to suppress EMT in breast cancer. Theranostics 2021; 11:8218-8233. [PMID: 34373738 PMCID: PMC8344017 DOI: 10.7150/thno.59280] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
Purpose: Functional loss of BRCA1 is associated with poorly differentiated and metastatic breast cancers that are enriched with cancer stem cells (CSCs). CSCs can be generated from carcinoma cells through an epithelial-mesenchymal transition (EMT) program. We and others have previously demonstrated that BRCA1 suppresses EMT and regulates the expression of multiple EMT-related transcription factors. However, the downstream mediators of BRCA1 function in EMT suppression remain elusive. Methods: Depletion of BRCA1 or GATA3 activates p18INK4C , a cell cycle inhibitor which inhibits mammary epithelial cell proliferation. We have therefore created genetically engineered mice with Brca1 or Gata3 loss in addition to deletion of p18INK4C , to rescue proliferative defects caused by deficiency of Brca1 or Gata3. By using these mutant mice along with human BRCA1 deficient as well as proficient breast cancer tissues and cells, we investigated and compared the role of Brca1 and Gata3 loss in the activation of EMT in breast cancers. Results: We discovered that BRCA1 and GATA3 expressions were positively correlated in human breast cancer. Depletion of BRCA1 stimulated methylation of GATA3 promoter thereby repressing GATA3 transcription. We developed Brca1 and Gata3 deficient mouse system. We found that Gata3 deficiency in mice induced poorly-differentiated mammary tumors with the activation of EMT and promoted tumor initiating and metastatic potential. Gata3 deficient mammary tumors phenocopied Brca1 deficient tumors in the induction of EMT under the same genetic background. Reconstitution of Gata3 in Brca1-deficient tumor cells activated mesenchymal-epithelial transition, suppressing tumor initiation and metastasis. Conclusions: Our finding, for the first time, demonstrates that GATA3 functions downstream of BRCA1 to suppress EMT in controlling mammary tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
| | - Li-Han Zhang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Chuying Wang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Chenglong Zheng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Jianping Sun
- Department of Mathematics and Statistics, University of North Carolina at Greensboro, Greensboro, NC 27402, USA
| | - Min Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, 730000, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136, USA
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen 518060, China
| |
Collapse
|
9
|
Tan JJ, Wang L, Mo TT, Dai YF, Lu J, Liu X, Chen HH, Tian WD, Li XP. Establishment of Immortalized Laryngeal Epithelial Cells Transfected with Bmi1. Cell Transplant 2021; 29:963689720908198. [PMID: 32249592 PMCID: PMC7444206 DOI: 10.1177/0963689720908198] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Primary laryngeal epithelial cells are essential to exploring the
mechanisms of laryngeal and voice disorders; however, they are
difficult to study and apply because of their limited life span. The
purpose of this study was to develop a stable and reliable in
vitro model for the comprehensive study of the
pathogenesis of laryngeal and voice diseases. The pLVTHM-Bmi1 plasmid
was constructed and used to immortalize primary laryngeal epithelial
cells by lentiviral infection. The expressions of Bmi1, human
telomerase reverse transcriptase (hTERT), p53, and pRB pathway
proteins were detected by western blotting. Functional characteristics
of the immortalized cell lines were verified by cell senescence
β-galactosidase staining, 5-ethynyl-2′-deoxyuridine cell proliferation
test, and flow cytometry. We successfully introduced Bmi into human
subglottic (hSG) cells and human ventricle (hV) cells. Both the human
immortalized subglottic Bmi1 (hSG-Bmi1) cell line and the human
immortalized ventricle Bmi1 (hV-Bmi1) cell line maintained normal
epithelial morphology and divided successfully after more than 20
culture passages. As Bmi1 was overexpressed in these cells, the
expression of human telomerase reverse transcriptase (hTERT) and
phosphorylated Rb increased while p16 and p21 decreased. Following
Bmi1-mediated immortalization, cell senescence decreased
significantly, and cell proliferation was accelerated. Tumor formation
was not observed for hSG, hV, or hSG-Bmi1, and hV-Bmi1 cells in nude
mice. hSG-Bmi1 cells dominated by stratified squamous epithelium and
hV-Bmi1 cells dominated by columnar cells were established. The new
cell lines lay a foundation for the study of the pathogenic mechanisms
of laryngeal and voice diseases.
Collapse
Affiliation(s)
- Jia-Jie Tan
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Both the authors are co-first authors and contributed equally to this article
| | - Lu Wang
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Otolaryngology, Head and Neck Surgery, Shenzhen University General Hospital, Shenzhen, Guangdong, China.,Both the authors are co-first authors and contributed equally to this article
| | - Ting-Ting Mo
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuan-Feng Dai
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Juan Lu
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiong Liu
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Huai-Hong Chen
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wen-Dong Tian
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiang-Ping Li
- Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Bai F, Liu S, Liu X, Hollern DP, Scott A, Wang C, Zhang L, Fan C, Fu L, Perou CM, Zhu WG, Pei XH. PDGFRβ is an essential therapeutic target for BRCA1-deficient mammary tumors. Breast Cancer Res 2021; 23:10. [PMID: 33478572 PMCID: PMC7819225 DOI: 10.1186/s13058-021-01387-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/03/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Basal-like breast cancers (BLBCs) are a leading cause of cancer death due to their capacity to metastasize and lack of effective therapies. More than half of BLBCs have a dysfunctional BRCA1. Although most BRCA1-deficient cancers respond to DNA-damaging agents, resistance and tumor recurrence remain a challenge to survival outcomes for BLBC patients. Additional therapies targeting the pathways aberrantly activated by BRCA1 deficiency are urgently needed. METHODS Most BRCA1-deficient BLBCs carry a dysfunctional INK4-RB pathway. Thus, we created genetically engineered mice with Brca1 loss and deletion of p16INK4A, or separately p18INK4C, to model the deficient INK4-RB signaling in human BLBC. By using these mutant mice and human BRCA1-deficient and proficient breast cancer tissues and cells, we tested if there exists a druggable target in BRCA1-deficient breast cancers. RESULTS Heterozygous germline or epithelium-specific deletion of Brca1 in p18INK4C- or p16INK4A-deficient mice activated Pdgfrβ signaling, induced epithelial-to-mesenchymal transition, and led to BLBCs. Confirming this role, targeted deletion of Pdgfrβ in Brca1-deficient tumor cells promoted cell death, induced mesenchymal-to-epithelial transition, and suppressed tumorigenesis. Importantly, we also found that pharmaceutical inhibition of Pdgfrβ and its downstream target Pkcα suppressed Brca1-deficient tumor initiation and progression and effectively killed BRCA1-deficient cancer cells. CONCLUSIONS Our work offers the first genetic and biochemical evidence that PDGFRβ-PKCα signaling is repressed by BRCA1, which establishes PDGFRβ-PKCα signaling as a therapeutic target for BRCA1-deficient breast cancers.
Collapse
Affiliation(s)
- Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Shiqin Liu
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Daniel P Hollern
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alexandria Scott
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Chuying Wang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lihan Zhang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, China
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
- Department of Pharmacology, Shenzhen University Health Science Center, Shenzhen, 518039, China
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China.
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA.
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| |
Collapse
|
11
|
Jo HG, Park C, Lee H, Kim GY, Keum YS, Hyun JW, Kwon TK, Choi YH, Hong SH. Inhibition of oxidative stress induced-cytotoxicity by coptisine in V79-4 Chinese hamster lung fibroblasts through the induction of Nrf-2 mediated HO-1 expression. Genes Genomics 2020; 43:17-31. [PMID: 33237503 DOI: 10.1007/s13258-020-01018-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 11/05/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Coptisine is a natural alkaloid compound and is known to have multiple beneficial effects including antioxidant activity. However, whether it can protect lung fibroblasts from oxidative damage has not been studied yet. OBJECTIVES To investigate the potential inhibitory effect of coptisine against oxidative stress in V79-4 lung fibroblast cells. METHODS V79-4 cells were treated with H2O2 (1 mM) in the presence or absence of coptisine (50 µg/ml), N-acetyl cysteine (NAC, 10 mM) or zinc protoporphyrin IX (ZnPP, 10 µM) for the indicated times. The alleviating effects of coptisine on cytotoxicity, cell cycle arrest, apoptosis, reactive oxygen species (ROS) production, DNA damage, mitochondrial dynamics, and inhibition of ATP production against H2O2 were investigated. Western blot analysis was used to analyze the expression levels of specific proteins. RESULTS Coptisine inhibited H2O2-induced cytotoxicity and DNA damage by blocking abnormal ROS generation. H2O2 treatment caused cell cycle arrest at the G2/M phase accompanied by increased expression of cyclin-dependent kinase (Cdk) inhibitor p21WAF1/CIP1 and decreased expression of cyclin B1 and cyclin A. However, these effects were attenuated in the presence of coptisine or NAC. Coptisine also prevented apoptosis by decreasing the rate of Bax/Bcl-2 expression in H2O2-stimulated cells and suppressing the loss of mitochondrial membrane potential and the cytosolic release of cytochrome c. In addition, the activation of nuclear factor-erythroid-2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) was markedly promoted by coptisine in the presence of H2O2. However, zinc protoporphyrin IX, a potent inhibitor of HO-1, attenuated the ROS scavenging and anti-apoptotic effects of coptisine. CONCLUSIONS Based on current data, we suggest that coptisine can be used as a potential treatment for oxidative stress-related lung disease.
Collapse
Affiliation(s)
- Hyeon-Gyun Jo
- Cheong-Choon Korean Medical Clinic, 47388, Busan, Republic of Korea.,Department of Biochemistry, Dong-eui University College of Korean Medicine, 47227, Busan, Republic of Korea
| | - Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-eui University, 47340, Busan, Republic of Korea
| | - Hyesook Lee
- Department of Biochemistry, Dong-eui University College of Korean Medicine, 47227, Busan, Republic of Korea.,Anti-Aging Research Center, Dong-eui University, 47340, Busan, Republic of Korea
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, 63243, Jeju, Republic of Korea
| | - Young-Sam Keum
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University, 10326, Goyang, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, 63243, Jeju, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, 42601, Daegu, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine, 47227, Busan, Republic of Korea. .,Anti-Aging Research Center, Dong-eui University, 47340, Busan, Republic of Korea.
| | - Su Hyun Hong
- Department of Biochemistry, Dong-eui University College of Korean Medicine, 47227, Busan, Republic of Korea. .,Anti-Aging Research Center, Dong-eui University, 47340, Busan, Republic of Korea.
| |
Collapse
|
12
|
Arredondo-Robles AV, Rodríguez-López KP, Ávila-Avilés RD. RETRACTED ARTICLE: Long non-coding RNAs in cervical cancer. J Appl Genet 2020; 61:405. [PMID: 31981186 DOI: 10.1007/s13353-020-00545-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/03/2020] [Accepted: 01/20/2020] [Indexed: 10/25/2022]
Affiliation(s)
- A V Arredondo-Robles
- Facultad de Ciencias, Universidad Autónoma del Estado de México, Campus El Cerrillo, Piedras Blancas, Carretera Toluca Kilómetro 15.5, C.P. 50200, Ixtlahuaca, Edo. de México, Mexico
| | - K P Rodríguez-López
- Facultad de Ciencias, Universidad Autónoma del Estado de México, Campus El Cerrillo, Piedras Blancas, Carretera Toluca Kilómetro 15.5, C.P. 50200, Ixtlahuaca, Edo. de México, Mexico
| | - R D Ávila-Avilés
- Facultad de Ciencias, Universidad Autónoma del Estado de México, Campus El Cerrillo, Piedras Blancas, Carretera Toluca Kilómetro 15.5, C.P. 50200, Ixtlahuaca, Edo. de México, Mexico. .,Departamento de Genética y Biología Molecular, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Av Instituto Politécnico Nacional, 2508, La Laguna Ticoman, C.P. 07360, Ciudad de México, Mexico.
| |
Collapse
|
13
|
Urbach A, Witte OW. Divide or Commit - Revisiting the Role of Cell Cycle Regulators in Adult Hippocampal Neurogenesis. Front Cell Dev Biol 2019; 7:55. [PMID: 31069222 PMCID: PMC6491688 DOI: 10.3389/fcell.2019.00055] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Accepted: 03/28/2019] [Indexed: 12/21/2022] Open
Abstract
The adult dentate gyrus continuously generates new neurons that endow the brain with increased plasticity, helping to cope with changing environmental and cognitive demands. The process leading to the birth of new neurons spans several precursor stages and is the result of a coordinated series of fate decisions, which are tightly controlled by extrinsic signals. Many of these signals act through modulation of cell cycle (CC) components, not only to drive proliferation, but also for linage commitment and differentiation. In this review, we provide a comprehensive overview on key CC components and regulators, with emphasis on G1 phase, and analyze their specific functions in precursor cells of the adult hippocampus. We explore their role for balancing quiescence versus self-renewal, which is essential to maintain a lifelong pool of neural stem cells while producing new neurons “on demand.” Finally, we discuss available evidence and controversies on the impact of CC/G1 length on proliferation versus differentiation decisions.
Collapse
Affiliation(s)
- Anja Urbach
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Jena, Germany
| |
Collapse
|
14
|
Frankell AM, Jammula S, Li X, Contino G, Killcoyne S, Abbas S, Perner J, Bower L, Devonshire G, Ococks E, Grehan N, Mok J, O'Donovan M, MacRae S, Eldridge MD, Tavaré S, Fitzgerald RC. The landscape of selection in 551 esophageal adenocarcinomas defines genomic biomarkers for the clinic. Nat Genet 2019; 51:506-516. [PMID: 30718927 PMCID: PMC6420087 DOI: 10.1038/s41588-018-0331-5] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 12/10/2018] [Indexed: 12/24/2022]
Abstract
Esophageal adenocarcinoma (EAC) is a poor-prognosis cancer type with rapidly rising incidence. Understanding of the genetic events driving EAC development is limited, and there are few molecular biomarkers for prognostication or therapeutics. Using a cohort of 551 genomically characterized EACs with matched RNA sequencing data, we discovered 77 EAC driver genes and 21 noncoding driver elements. We identified a mean of 4.4 driver events per tumor, which were derived more commonly from mutations than copy number alterations, and compared the prevelence of these mutations to the exome-wide mutational excess calculated using non-synonymous to synonymous mutation ratios (dN/dS). We observed mutual exclusivity or co-occurrence of events within and between several dysregulated EAC pathways, a result suggestive of strong functional relationships. Indicators of poor prognosis (SMAD4 and GATA4) were verified in independent cohorts with significant predictive value. Over 50% of EACs contained sensitizing events for CDK4 and CDK6 inhibitors, which were highly correlated with clinically relevant sensitivity in a panel of EAC cell lines and organoids.
Collapse
Affiliation(s)
- Alexander M Frankell
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK
| | - SriGanesh Jammula
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Xiaodun Li
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK
| | - Gianmarco Contino
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK
| | - Sarah Killcoyne
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK
| | - Sujath Abbas
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK
| | - Juliane Perner
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Lawrence Bower
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Ginny Devonshire
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Emma Ococks
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK
| | - Nicola Grehan
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK
| | - James Mok
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK
| | | | - Shona MacRae
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK
| | - Matthew D Eldridge
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Simon Tavaré
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Rebecca C Fitzgerald
- MRC cancer unit, Hutchison/MRC research Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
15
|
Differential expression of tumor-associated genes and altered gut microbiome with decreased Akkermansia muciniphila confer a tumor-preventive microenvironment in intestinal epithelial Pten-deficient mice. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3746-3758. [PMID: 30292635 DOI: 10.1016/j.bbadis.2018.10.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 09/22/2018] [Accepted: 10/02/2018] [Indexed: 12/15/2022]
Abstract
Phosphatase and tensin homolog (Pten) antagonizes PI3K-Akt signaling; therefore, Pten impairment causes tumorigenesis. However, the correlation between Pten deficiency and colon cancer has remained elusive due to numerous opposite observations. To study this correlation, we examined whether Pten deficiency in intestinal epithelial cells (IECs) induces tumorigenesis. With mucosal biopsies of human colon cancer and normal colon, Pten mRNA was evaluated by quantitative PCR. Using IEC-specific Pten knockout mice (PtenΔIEC/ΔIEC), we examined the mitotic activity of IECs; and PtenΔIEC/ΔIEC; Apcmin/+ mice were generated by combining PtenΔIEC/ΔIEC with Apcmin/+ mice. Tumor-associated gene was evaluated by micro-array analysis. Fecal microbiome was analyzed through 16S rRNA gene sequencing. We found that Pten mRNA level was reduced in human colon cancer relative to normal tissues. Augmented chromatids, increased Ki-67 and PCNA expression, and enhanced Akt activation were identified in IECs of PtenΔIEC/ΔIEC mice compared to Pten+/+ littermate. Combining PtenΔIEC/ΔIEC with Apcmin/+ condition caused rapid and aggressive intestinal tumorigenesis. However, PtenΔIEC/ΔIEC mice did not develop any tumors. While maintaining the tumor-driving potential, these data indicated that IEC-Pten deficiency alone did not induce tumorigenesis in mice. Furthermore, the expression of tumor-promoting and tumor-suppressing genes was decreased and increased, respectively, in the intestine of PtenΔIEC/ΔIEC mice compared to controls. The abundance of Akkermansia muciniphila, capable of inducing chronic intestinal inflammation, was diminished in PtenΔIEC/ΔIEC mice compared to controls. These findings suggested that altered tumor-associated gene expression and changed gut microbiota shape a tumor-preventive microenvironment to counteract the tumor-driving potential, leading to the tumor prevention in PtenΔIEC/ΔIEC mice.
Collapse
|
16
|
Wang C, Bai F, Zhang LH, Scott A, Li E, Pei XH. Estrogen promotes estrogen receptor negative BRCA1-deficient tumor initiation and progression. Breast Cancer Res 2018; 20:74. [PMID: 29996906 PMCID: PMC6042319 DOI: 10.1186/s13058-018-0996-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/30/2018] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Estrogen promotes breast cancer development and progression mainly through estrogen receptor (ER). However, blockage of estrogen production or action prevents development of and suppresses progression of ER-negative breast cancers. How estrogen promotes ER-negative breast cancer development and progression is poorly understood. We previously discovered that deletion of cell cycle inhibitors p16Ink4a (p16) or p18Ink4c (p18) is required for development of Brca1-deficient basal-like mammary tumors, and that mice lacking p18 develop luminal-type mammary tumors. METHODS A genetic model system with three mouse strains, one that develops ER-positive mammary tumors (p18 single deletion) and the others that develop ER-negative tumors (p16;Brca1 and p18;Brca1 compound deletion), human BRCA1 mutant breast cancer patient-derived xenografts, and human BRCA1-deficient and BRCA1-proficient breast cancer cells were used to determine the role of estrogen in activating epithelial-mesenchymal transition (EMT), stimulating cell proliferation, and promoting ER-negative mammary tumor initiation and metastasis. RESULTS Estrogen stimulated the proliferation and tumor-initiating potential of both ER-positive Brca1-proficient and ER-negative Brca1-deficient tumor cells. Estrogen activated EMT in a subset of Brca1-deficient mammary tumor cells that maintained epithelial features, and enhanced the number of cancer stem cells, promoting tumor progression and metastasis. Estrogen activated EMT independent of ER in Brca1-deficient, but not Brca1-proficient, tumor cells. Estrogen activated the AKT pathway in BRCA1-deficient tumor cells independent of ER, and pharmaceutical inhibition of AKT activity suppressed EMT and cell proliferation preventing BRCA1 deficient tumor progression. CONCLUSIONS This study reveals for the first time that estrogen promotes BRCA1-deficient tumor initiation and progression by stimulation of cell proliferation and activation of EMT, which are dependent on AKT activation and independent of ER.
Collapse
Affiliation(s)
- Chuying Wang
- Department of Medical Oncology, The First Affiliated hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 People’s Republic of China
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Feng Bai
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Li-han Zhang
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Alexandria Scott
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
| | - Enxiao Li
- Department of Medical Oncology, The First Affiliated hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 People’s Republic of China
| | - Xin-Hai Pei
- Molecular Oncology Program, Division of Surgical Oncology, Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL 33136 USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136 USA
| |
Collapse
|
17
|
Liu S, Chan HL, Bai F, Ma J, Scott A, Robbins DJ, Capobianco AJ, Zhu P, Pei XH. Gata3 restrains B cell proliferation and cooperates with p18INK4c to repress B cell lymphomagenesis. Oncotarget 2018; 7:64007-64020. [PMID: 27588406 PMCID: PMC5325421 DOI: 10.18632/oncotarget.11746] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/24/2016] [Indexed: 12/13/2022] Open
Abstract
GATA3, a lineage specifier, controls lymphoid cell differentiation and its function in T cell commitment and development has been extensively studied. GATA3 promotes T cell specification by repressing B cell potential in pro T cells and decreased GATA3 expression is essential for early B cell commitment. Inherited genetic variation in GATA3 has been associated with lymphoma susceptibility. However, it remains elusive how the loss of function of GATA3 promotes B cell development and induces B cell lymphomas. In this study, we found that haploid loss of Gata3 by heterozygous germline deletion increased B cell populations in the bone marrow (BM) and spleen, and decreased CD4 T cell populations in the thymus, confirming that Gata3 promotes T and suppresses B cell development. We discovered that haploid loss of Gata3 reduced thymocyte proliferation with induction of p18Ink4c (p18), an inhibitor of CDK4 and CDK6, but enhanced B cell proliferation in the BM and spleen independent of p18. Loss of p18 partially restored Gata3 deficient thymocyte proliferation, but further stimulated Gata3 deficient B cell proliferation in the BM and spleen. Furthermore, we discovered that haploid loss of Gata3 in p18 deficient mice led to the development of B cell lymphomas that were capable of rapidly regenerating tumors when transplanted into immunocompromised mice. These results indicate that Gata3 deficiency promotes B cell differentiation and proliferation, and cooperates with p18 loss to induce B cell lymphomas. This study, for the first time, reveals that Gata3 is a tumor suppressor specifically in B cell lymphomagenesis.
Collapse
Affiliation(s)
- Shiqin Liu
- Department of Hematology, Peking University First Hospital, Beijing, 100034, China.,Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Ho Lam Chan
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Feng Bai
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Jinshan Ma
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,Xinjiang Uigur Autonomous Region People's Hospital, Xinjiang, 830001, China
| | - Alexandria Scott
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - David J Robbins
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Anthony J Capobianco
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, FL 33136, Miami
| | - Ping Zhu
- Department of Hematology, Peking University First Hospital, Beijing, 100034, China
| | - Xin-Hai Pei
- Molecular Oncology Program, Division of Surgical Oncology, Department of Surgery, Miller School of Medicine, University of Miami, FL 33136, Miami.,The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, FL 33136, Miami.,Sylvester Cancer Center, Miller School of Medicine, University of Miami, FL 33136, Miami
| |
Collapse
|
18
|
Oak C, Khalifa AO, Isali I, Bhaskaran N, Walker E, Shukla S. Diosmetin suppresses human prostate cancer cell proliferation through the induction of apoptosis and cell cycle arrest. Int J Oncol 2018; 53:835-843. [PMID: 29767250 PMCID: PMC6017185 DOI: 10.3892/ijo.2018.4407] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/30/2018] [Indexed: 12/15/2022] Open
Abstract
Diosmetin, a plant flavonoid, has been shown to exert promising effects on prostate cancer cells as an anti‑proliferative and anticancer agent. In this study, using western blot analysis for protein expression and flow cytometry for cell cycle analysis, we determined that the treatment of the LNCaP and PC‑3 prostate cancer cells with diosmetin resulted in a marked decrease in cyclin D1, Cdk2 and Cdk4 expression levels (these proteins remain active in the G0‑G1 phases of the cell cycle). These changes were accompanied by a decrease in c-Myc and Bcl-2 expression, and by an increase in Bax, p27Kip1 and FOXO3a protein expression, which suggests the potential modulatory effects of diosmetin on protein transcription. The treatment of prostate cancer cells with diosmetin set in motion an apoptotic machinery by inhibiting X-linked inhibitor of apoptosis (XIAP) and increasing cleaved PARP and cleaved caspase-3 expression levels. On the whole, the findings of this study provide an in-depth analysis of the molecular mechanisms responsible for the regulatory effects of diosmetin on key molecules that perturb the cell cycle to inhibit cell growth, and suggest that diosmetin may prove to be an effective anticancer agent for use in the treatment of prostate cancer in the future.
Collapse
Affiliation(s)
- Christine Oak
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Ahmad O Khalifa
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Ilaha Isali
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Natarajan Bhaskaran
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Ethan Walker
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sanjeev Shukla
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
19
|
Yoon N, Yoon G, Park CK, Kim HS. Stromal p16 expression is significantly increased in malignant ovarian neoplasms. Oncotarget 2018; 7:64665-64673. [PMID: 27572321 PMCID: PMC5323106 DOI: 10.18632/oncotarget.11660] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 08/24/2016] [Indexed: 12/21/2022] Open
Abstract
Alterations in p16 protein expression have been reported to be associated with tumor development and progression. However, p16 expression status in the peritumoral stroma has been rarely investigated. We investigated the stromal p16 expression in ovarian neoplasms using immunohistochemistry, and differences in the expression status depending on the degree of malignancy and histological type were analyzed. This study included 24, 21, and 46 cases of benign, borderline, and malignant ovarian lesions, respectively, of which 29, 25, and 32 cases were serous, mucinous, and endometriosis-associated lesions. Most benign lesions showed negative or weak expression, whereas borderline lesions showed focal, moderate expression. Malignant lesions showed markedly elevated stromal p16 expression compared with benign or borderline lesions. There were significant differences in stromal p16 expression between benign and borderline lesions (P < 0.001) and between borderline and malignant lesions (P < 0.001). These significances remained when analysis was performed based on lesion classification as serous, mucinous, and endometriosis-associated. In contrast, differences in stromal p16 expression among the histological types were not significant. Stromal p16 expression in ovarian neoplasms was absent or weak in benign and focal, moderate in borderline lesions, whereas malignant lesions exhibited diffuse, moderate-to-strong p16 immunoreactivity. Our observations suggest that stromal p16 expression is involved in the development of ovarian carcinoma. Further studies are necessary to confirm our preliminary results.
Collapse
Affiliation(s)
- Nara Yoon
- Department of Pathology, The Catholic University of Korea Incheon St. Mary's Hospital, Incheon, Republic of Korea
| | - Gun Yoon
- Shinsegae Women's Hospital, Daegu, Republic of Korea
| | - Cheol Keun Park
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun-Soo Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
20
|
Yoon G, Koh CW, Yoon N, Kim JY, Kim HS. Stromal p16 expression is significantly increased in endometrial carcinoma. Oncotarget 2018; 8:4826-4836. [PMID: 27902476 PMCID: PMC5354874 DOI: 10.18632/oncotarget.13594] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/16/2016] [Indexed: 11/25/2022] Open
Abstract
p16 is a negative regulator of cell proliferation and is considered a tumor suppressor protein. Alterations in p16 protein expression are associated with tumor development and progression. However, the p16 expression status in the peritumoral stroma has not been investigated in the endometrium. Therefore, we evaluated stromal p16 expression in different types of endometrial lesions using immunohistochemistry. Differences in the p16 expression status according to the degree of malignancy and histological type were analyzed. This study included 62, 26, and 36 cases of benign, precancerous, and malignant endometrial lesions, respectively. Most benign lesions showed negative or weak expression, whereas precancerous lesions showed a variable degree of staining proportion and intensity. Atypical hyperplasia/endometrial intraepithelial neoplasia (AH/EIN) and serous endometrial intraepithelial carcinoma (SEIC) had significantly higher stromal p16 expression levels than benign lesions. Endometrioid carcinoma (EC), serous carcinoma (SC), and carcinosarcoma showed significantly elevated stromal p16 expression levels compared with benign and precancerous lesions. In addition, there were significant differences in stromal p16 expression between AH/EIN and SEIC and between EC and SC. In contrast, differences in stromal p16 expression among nonpathological endometrium, atrophic endometrium, endometrial polyp, and hyperplasia without atypia were not statistically significant. Our observations suggest that stromal p16 expression is involved in the development and progression of endometrial carcinoma, and raise the possibility that p16 overexpression in the peritumoral stroma is associated with aggressive oncogenic behavior of endometrial SC.
Collapse
Affiliation(s)
- Gun Yoon
- Shinsegae Women's Hospital, Daegu, Republic of Korea
| | - Chang Won Koh
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Nara Yoon
- Department of Pathology, The Catholic University of Korea Incheon St. Mary's Hospital, Incheon, Republic of Korea
| | - Ji-Ye Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun-Soo Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
21
|
Aasrum M, Thoresen GH, Christoffersen T, Brusevold IJ. p38 differentially regulates ERK, p21, and mitogenic signalling in two pancreatic carcinoma cell lines. J Cell Commun Signal 2018; 12:699-707. [PMID: 29380233 DOI: 10.1007/s12079-017-0444-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/18/2017] [Indexed: 01/24/2023] Open
Abstract
Whereas the p38 MAP kinase has largely been associated with anti-proliferative functions, several observations have indicated that it may also have positive effects on proliferation. In hepatocytes, we have found that p38 has opposing effects on DNA synthesis when activated by EGF and HGF. Here we have studied the function of p38 in EGF- and HGF-induced DNA synthesis in the two pancreatic carcinoma cell lines AsPC-1 and Panc-1. In Panc-1 cells, the MEK inhibitor PD98059 reduced EGF- and HGF-induced DNA synthesis, while the p38 inhibitor SB203580 strongly increased the basal DNA synthesis and reduced expression of the cyclin-dependent kinase inhibitor (CDKI) p21. In contrast, in AsPC-1 cells, EGF- and HGF-induced DNA synthesis was not significantly reduced by PD98059 but was inhibited by SB203580. Treatment with SB203580 amplified the sustained ERK phosphorylation induced by these growth factors and caused a marked upregulation of the expression of p21, which could be blocked by PD98059. These results suggest that while DNA synthesis in Panc-1 cells is enhanced by ERK and strongly suppressed by p38, in AsPC-1 cells, p38 exerts a pro-mitogenic effect through MEK/ERK-dependent downregulation of p21. Thus, p38 may have suppressive or stimulatory effects on proliferation depending on the cell type, due to differential cross-talk between the p38 and MEK/ERK pathways.
Collapse
Affiliation(s)
- Monica Aasrum
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, P.O. Box 1057, Blindern, 0316, Oslo, Norway.
| | - G Hege Thoresen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, P.O. Box 1057, Blindern, 0316, Oslo, Norway
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - Thoralf Christoffersen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, P.O. Box 1057, Blindern, 0316, Oslo, Norway
| | - Ingvild J Brusevold
- Department of Paediatric Dentistry and Behavioural Science, Faculty of Dentistry, University of Oslo, Oslo, Norway
| |
Collapse
|
22
|
Vengoechea J, Tallo C. A germline deletion of 9p21.3 presenting as familial melanoma, astrocytoma and breast cancer: clinical and genetic counselling challenges. J Med Genet 2017; 54:682-684. [DOI: 10.1136/jmedgenet-2017-104690] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 04/24/2017] [Accepted: 05/26/2017] [Indexed: 01/07/2023]
|
23
|
Terzibasi-Tozzini E, Martinez-Nicolas A, Lucas-Sánchez A. The clock is ticking. Ageing of the circadian system: From physiology to cell cycle. Semin Cell Dev Biol 2017. [PMID: 28630025 DOI: 10.1016/j.semcdb.2017.06.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The circadian system is the responsible to organise the internal temporal order in relation to the environment of every process of the organisms producing the circadian rhythms. These rhythms have a fixed phase relationship among them and with the environment in order to optimise the available energy and resources. From a cellular level, circadian rhythms are controlled by genetic positive and negative auto-regulated transcriptional and translational feedback loops, which generate 24h rhythms in mRNA and protein levels of the clock components. It has been described about 10% of the genome is controlled by clock genes, with special relevance, due to its implications, to the cell cycle. Ageing is a deleterious process which affects all the organisms' structures including circadian system. The circadian system's ageing may produce a disorganisation among the circadian rhythms, arrhythmicity and, even, disconnection from the environment, resulting in a detrimental situation to the organism. In addition, some environmental conditions can produce circadian disruption, also called chronodisruption, which may produce many pathologies including accelerated ageing. Finally, some strategies to prevent, palliate or counteract chronodisruption effects have been proposed to enhance the circadian system, also called chronoenhancement. This review tries to gather recent advances in the chronobiology of the ageing process, including cell cycle, neurogenesis process and physiology.
Collapse
Affiliation(s)
| | - Antonio Martinez-Nicolas
- Department of Physiology, Faculty of Biology, University of Murcia, Campus Mare Nostrum, IUIE. IMIB-Arrixaca, Murcia, Spain; Ciber Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Alejandro Lucas-Sánchez
- Department of Physiology, Faculty of Biology, University of Murcia, Campus Mare Nostrum, IUIE. IMIB-Arrixaca, Murcia, Spain; Ciber Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain.
| |
Collapse
|
24
|
Morris-Hanon O, Furmento VA, Rodríguez-Varela MS, Mucci S, Fernandez-Espinosa DD, Romorini L, Sevlever GE, Scassa ME, Videla-Richardson GA. The Cell Cycle Inhibitors p21 Cip1 and p27 Kip1 Control Proliferation but Enhance DNA Damage Resistance of Glioma Stem Cells. Neoplasia 2017; 19:519-529. [PMID: 28582703 PMCID: PMC5458648 DOI: 10.1016/j.neo.2017.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/28/2017] [Accepted: 04/03/2017] [Indexed: 01/06/2023] Open
Abstract
High-grade gliomas are the most prevalent and lethal primary brain tumors. They display a hierarchical arrangement with a population of self-renewing and highly tumorigenic cells called cancer stem cells. These cells are thought to be responsible for tumor recurrence, which make them main candidates for targeted therapies. Unbridled cell cycle progression may explain the selective sensitivity of some cancer cells to treatments. The members of the Cip/Kip family p21Cip1 and p27Kip1 were initially considered as tumor suppressors based on their ability to block proliferation. However, they are currently looked at as proteins with dual roles in cancer: one as tumor suppressor and the other as oncogene. Therefore, the aim of this study was to determine the functions of these cell cycle inhibitors in five patient-derived glioma stem cell–enriched cell lines. We found that these proteins are functional in glioma stem cells. They negatively regulate cell cycle progression both in unstressed conditions and in response to genotoxic stress. In addition, p27Kip1 is upregulated in nutrient-restricted and differentiating cells, suggesting that this Cip/Kip is a mediator of antimitogenic signals in glioma cells. Importantly, the lack of these proteins impairs cell cycle halt in response to genotoxic agents, rendering cells more vulnerable to DNA damage. For these reasons, these proteins may operate both as tumor suppressors, limiting cell proliferation, and as oncogenes, conferring cell resistance to DNA damage. Thus, deepening our knowledge on the biological functions of these Cip/Kips may shed light on how some cancer cells develop drug resistance.
Collapse
Affiliation(s)
- Olivia Morris-Hanon
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Ruta 9, Km 52.5, B1625XAF, Escobar, Provincia de Buenos Aires, Argentina.
| | - Verónica Alejandra Furmento
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Ruta 9, Km 52.5, B1625XAF, Escobar, Provincia de Buenos Aires, Argentina.
| | - María Soledad Rodríguez-Varela
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Ruta 9, Km 52.5, B1625XAF, Escobar, Provincia de Buenos Aires, Argentina.
| | - Sofía Mucci
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Ruta 9, Km 52.5, B1625XAF, Escobar, Provincia de Buenos Aires, Argentina.
| | - Damián Darío Fernandez-Espinosa
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Ruta 9, Km 52.5, B1625XAF, Escobar, Provincia de Buenos Aires, Argentina.
| | - Leonardo Romorini
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Ruta 9, Km 52.5, B1625XAF, Escobar, Provincia de Buenos Aires, Argentina.
| | - Gustavo Emilio Sevlever
- Laboratorio de Neuropatología, Departamento de Neuropatología y Biología Molecular, Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Montañeses 2325, C1428AQK, Buenos Aires, Argentina.
| | - María Elida Scassa
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Ruta 9, Km 52.5, B1625XAF, Escobar, Provincia de Buenos Aires, Argentina.
| | - Guillermo Agustín Videla-Richardson
- Laboratorio de Investigación aplicada a Neurociencias (LIAN), Fundación para la Lucha contra las Enfermedades Neurológicas de la Infancia (FLENI), Ruta 9, Km 52.5, B1625XAF, Escobar, Provincia de Buenos Aires, Argentina.
| |
Collapse
|
25
|
MARK2 inhibits the growth of HeLa cells through AMPK and reverses epithelial-mesenchymal transition. Oncol Rep 2017; 38:237-244. [PMID: 28560405 DOI: 10.3892/or.2017.5686] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 05/03/2017] [Indexed: 11/05/2022] Open
Abstract
Microtubule affinity-regulating kinases (MARKs; MARK1, MARK2, MARK3 and MARK4) act directly downstream of LKB1, the multitasking tumor-suppressor kinase, and thereby mediate its biological effects. Current understanding of the function of MARKs is greatly restricted to regulation of cell polarity. However, whether or how MARKs contribute to cellular growth control remains largely unknown. In the present study, we utilized an inducible lentiviral expression system that allows rapid MARK expression in LKB1-deficient HeLa cells, and characterized additional functions of MARKs: overexpression of MARK2 in HeLa cells resulted in a decrease in cell growth, inhibition of colony formation and arrest in G1 cell cycle phase, with AMPK as the putative downstream effector upregulating the expression of p21 and p16. MARK2 was found to play a role in F-actin reorganization and to contribute to reversal of epithelial‑mesenchymal transition (EMT) as exemplified in the case of HeLa cells that exhibited phenotypic changes, reduced cell migration and invasion. Our findings unveil the coordinated regulation of cell growth and EMT mediated by MARK2, and also provide new insights into the mechanisms underlying the anti-metastatic activity of MARK2.
Collapse
|
26
|
Scott A, Bai F, Chan HL, Liu S, Slingerland JM, Robbins DJ, Capobianco AJ, Pei XH. p16 loss rescues functional decline of Brca1-deficient mammary stem cells. Cell Cycle 2017; 16:759-764. [PMID: 28278054 PMCID: PMC5405722 DOI: 10.1080/15384101.2017.1295185] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/19/2017] [Accepted: 02/08/2017] [Indexed: 01/04/2023] Open
Abstract
Recent evidence indicates that the accumulation of endogenous DNA damage can induce senescence and limit the function of adult stem cells. It remains elusive whether deficiency in DNA damage repair is associated with the functional alteration of mammary stem cells. In this article, we reported that senescence was induced in mammary epithelial cells during aging along with increased expression of p16Ink4a (p16), an inhibitor of CDK4 and CKD6. Loss of p16 abrogated the age-induced senescence in mammary epithelial cells and significantly increased mammary stem cell function. We showed that loss of Brca1, a tumor suppressor that functions in DNA damage repair, in the mammary epithelium induced senescence with induction of p16 and a decline of stem cell function, which was rescued by p16 loss. These data not only answer the question as to whether deficiency in DNA damage repair is associated with the functional decline of mammary stem cells, but also identify the role of p16 in suppressing Brca1-deficient mammary stem cell function.
Collapse
Affiliation(s)
- Alexandria Scott
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Feng Bai
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Ho Lam Chan
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Shiqin Liu
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Joyce M. Slingerland
- Braman Family Breast Cancer Institute, Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - David J. Robbins
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Anthony J. Capobianco
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Xin-Hai Pei
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
27
|
Scott A, Bai F, Chan HL, Liu S, Ma J, Slingerland JM, Robbins DJ, Capobianco AJ, Pei XH. p16INK4a suppresses BRCA1-deficient mammary tumorigenesis. Oncotarget 2016; 7:84496-84507. [PMID: 27811360 PMCID: PMC5356676 DOI: 10.18632/oncotarget.13015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 10/25/2016] [Indexed: 11/25/2022] Open
Abstract
Senescence prevents the proliferation of genomically damaged, but otherwise replication competent cells at risk of neoplastic transformation. p16INK4A (p16), an inhibitor of CDK4 and CDK6, plays a critical role in controlling cellular senescence in multiple organs. Functional inactivation of p16 by gene mutation and promoter methylation is frequently detected in human breast cancers. However, deleting p16 in mice or targeting DNA methylation within the murine p16 promoter does not result in mammary tumorigenesis. How loss of p16 contributes to mammary tumorigenesis in vivo is not fully understood.In this article, we reported that disruption of Brca1 in the mammary epithelium resulted in premature senescence that was rescued by p16 loss. We found that p16 loss transformed Brca1-deficient mammary epithelial cells and induced mammary tumors, though p16 loss alone was not sufficient to induce mammary tumorigenesis. We demonstrated that loss of both p16 and Brca1 led to metastatic, basal-like, mammary tumors with the induction of EMT and an enrichment of tumor initiating cells. We discovered that promoter methylation silenced p16 expression in most of the tumors developed in mice heterozygous for p16 and lacking Brca1. These data not only identified the function of p16 in suppressing BRCA1-deficient mammary tumorigenesis, but also revealed a collaborative effect of genetic mutation of p16 and epigenetic silencing of its transcription in promoting tumorigenesis. To the best of our knowledge, this is the first genetic evidence directly showing that p16 which is frequently deleted and inactivated in human breast cancers, collaborates with Brca1 controlling mammary tumorigenesis.
Collapse
MESH Headings
- Animals
- BRCA1 Protein/genetics
- BRCA1 Protein/metabolism
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cells, Cultured
- Cyclin-Dependent Kinase Inhibitor p16/genetics
- Cyclin-Dependent Kinase Inhibitor p16/metabolism
- DNA Methylation
- Epithelial Cells/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Mammary Neoplasms, Animal/genetics
- Mammary Neoplasms, Animal/metabolism
- Mammary Neoplasms, Animal/pathology
- Mice, Knockout
- Mice, Transgenic
- Promoter Regions, Genetic/genetics
Collapse
Affiliation(s)
- Alexandria Scott
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Feng Bai
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Ho Lam Chan
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Shiqin Liu
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Jinshan Ma
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Joyce M Slingerland
- Braman Family Breast Cancer Institute, Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - David J. Robbins
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Anthony J. Capobianco
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Xin-Hai Pei
- Molecular Oncology Program, Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- The Sheila and David Fuente Graduate Program in Cancer Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
- Sylvester Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
28
|
Wang Y, Cornett A, King FJ, Mao Y, Nigsch F, Paris CG, McAllister G, Jenkins JL. Evidence-Based and Quantitative Prioritization of Tool Compounds in Phenotypic Drug Discovery. Cell Chem Biol 2016; 23:862-874. [PMID: 27427232 DOI: 10.1016/j.chembiol.2016.05.016] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 04/29/2016] [Accepted: 05/13/2016] [Indexed: 01/07/2023]
Abstract
The use of potent and selective chemical tools with well-defined targets can help elucidate biological processes driving phenotypes in phenotypic screens. However, identification of selective compounds en masse to create targeted screening sets is non-trivial. A systematic approach is needed to prioritize probes, which prevents the repeated use of published but unselective compounds. Here we performed a meta-analysis of integrated large-scale, heterogeneous bioactivity data to create an evidence-based, quantitative metric to systematically rank tool compounds for targets. Our tool score (TS) was then tested on hundreds of compounds by assessing their activity profiles in a panel of 41 cell-based pathway assays. We demonstrate that high-TS tools show more reliably selective phenotypic profiles than lower-TS compounds. Additionally we highlight frequently tested compounds that are non-selective tools and distinguish target family polypharmacology from cross-family promiscuity. TS can therefore be used to prioritize compounds from heterogeneous databases for phenotypic screening.
Collapse
Affiliation(s)
- Yuan Wang
- Novartis Institutes for BioMedical Research Inc., 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Allen Cornett
- Novartis Institutes for BioMedical Research Inc., 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Fred J King
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA
| | - Yi Mao
- Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Florian Nigsch
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Novartis Campus, Basel 4056, Switzerland
| | - C Gregory Paris
- Novartis Institutes for BioMedical Research Inc., 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Gregory McAllister
- Novartis Institutes for BioMedical Research Inc., 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Jeremy L Jenkins
- Novartis Institutes for BioMedical Research Inc., 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
29
|
Ma Q, Grati M, Bai F, Pei J, Pei XH, Liu X. Rescue from early-onset hearing loss in a mouse model lacking the cyclin-dependent kinase inhibitor p19Ink4d. Cell Death Dis 2016; 7:e2131. [PMID: 26962681 PMCID: PMC4823939 DOI: 10.1038/cddis.2016.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Q Ma
- Department of Otolaryngology (D-48), University of Miami Miller School of Medicine, Miami, FL, USA
| | - M Grati
- Department of Otolaryngology (D-48), University of Miami Miller School of Medicine, Miami, FL, USA
| | - F Bai
- Molecular Oncology Program, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - J Pei
- Department of Otolaryngology (D-48), University of Miami Miller School of Medicine, Miami, FL, USA
| | - X-H Pei
- Molecular Oncology Program, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - X Liu
- Department of Otolaryngology (D-48), University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
30
|
Brookes S, Gagrica S, Sanij E, Rowe J, Gregory FJ, Hara E, Peters G. Evidence for a CDK4-dependent checkpoint in a conditional model of cellular senescence. Cell Cycle 2016; 14:1164-73. [PMID: 25695870 PMCID: PMC4613988 DOI: 10.1080/15384101.2015.1010866] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Cellular senescence, the stable cell cycle arrest elicited by various forms of stress, is an important facet of tumor suppression. Although much is known about the key players in the implementation of senescence, including the pRb and p53 axes and the cyclin dependent kinase inhibitors p16INK4a and p21CIP1, many details remain unresolved. In studying conditional senescence in human fibroblasts that express a temperature sensitive SV40 large T-antigen (T-Ag), we uncovered an unexpected role for CDK4. At the permissive temperature, where pRb and p53 are functionally compromised by T-Ag, cyclin D-CDK4 complexes are disrupted by the high p16INK4a levels and reduced expression of p21CIP1. In cells arrested at the non-permissive temperature, p21CIP1 promotes reassembly of cyclin D-CDK4 yet pRb is in a hypo-phosphorylated state, consistent with cell cycle arrest. In exploring whether the reassembled cyclin D-CDK4-p21 complexes are functional, we found that shRNA-mediated knockdown or chemical inhibition of CDK4 prevented the increase in cell size associated with the senescent phenotype by allowing the cells to arrest in G1 rather than G2/M. The data point to a role for CDK4 kinase activity in a G2 checkpoint that contributes to senescence.
Collapse
Key Words
- BrdU, bromodeoxyuridine
- CDK, cyclin dependent kinase
- CDK4
- FACS, fluorescence actvated cell sorting
- HFs, human fibroblasts
- PI, propidium iodide
- SA-βgal, senescence-associated β-galactosidase activity
- SV40 T-antigen
- SV40, simian virus 40
- TERT, telomerase reverse transcriptase
- human fibroblasts
- p16INK4a
- p21CIP1
- p53
- pRb, retinoblastoma protein
- retinoblastoma protein
- senescence
- shRNA, short-hairpin RNA
- ts, temperature sensitive
Collapse
Affiliation(s)
- Sharon Brookes
- a Cancer Research-UK London Research Institute ; London , UK
| | | | | | | | | | | | | |
Collapse
|
31
|
Kurimchak A, Graña X. PP2A: more than a reset switch to activate pRB proteins during the cell cycle and in response to signaling cues. Cell Cycle 2015; 14:18-30. [PMID: 25483052 PMCID: PMC4612414 DOI: 10.4161/15384101.2014.985069] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
In their active hypophosphorylated state, members of the retinoblastoma family of pocket proteins negatively regulate cell cycle progression at least in part by repressing expression of E2F-dependent genes. Mitogen-dependent activation of G1 and G1/S Cyclin Dependent Kinases (CDKs) results in coordinated hyperphosphorylation and inactivation of these proteins, which no longer bind and repress E2Fs. S and G2/M CDKs maintain pocket protein hyperphosphorylated through the end of mitosis. The inactivating action of inducible CDKs is opposed by the Ser/Thr protein phosphatases PP2A and PP1. Various trimeric PP2A holoenzymes have been implicated in dephosphorylation of pocket proteins in response to specific cellular signals and stresses or as part of an equilibrium with CDKs throughout the cell cycle. PP1 has specifically been implicated in dephosphorylation of pRB in late mitosis and early G1. This review is particularly focused on the emerging role of PP2A as a major hub for integration of growth suppressor signals that require rapid inactivation of pocket proteins. Of note, activation of particular PP2A holoenzymes triggers differential activation of pocket proteins in the presence of active CDKs.
Collapse
Affiliation(s)
- Alison Kurimchak
- a Fels Institute for Cancer Research and Molecular Biology and Department of Biochemistry; Temple University School of Medicine ; Philadelphia , PA USA
| | | |
Collapse
|
32
|
Jiang X, Shan A, Su Y, Cheng Y, Gu W, Wang W, Ning G, Cao Y. miR-144/451 Promote Cell Proliferation via Targeting PTEN/AKT Pathway in Insulinomas. Endocrinology 2015; 156:2429-39. [PMID: 25919186 DOI: 10.1210/en.2014-1966] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Insulinoma is the main type of functional pancreatic neuroendocrine tumors. The functional microRNAs (miRNAs) regulating tumor growth and progression in insulinomas are still unknown. We conducted the miRNA expression profile analysis using miRNA quantitative RT-PCR array and identified 114 differentially expressed miRNAs in human insulinomas compared with normal pancreatic islets. Forty-one differentially expressed miRNAs belonged to 7 miRNA families, and 28 miRNAs in 3 of the families localized in the epigenetically regulated imprinted chromosome 14q32 region. We validated the most significant differentially expressed miRNA cluster miR-144/451 in another 8 human normal islet samples and 25 insulinomas. Our data showed that the overexpression of miR-144/451 in mouse pancreatic β-cells promoted cell proliferation by targeting the β-cell regulator phosphatase and tensin homolog deleted on chromosome ten/v-akt murine thymoma viral oncogene homolog pathway and cyclin-dependent kinase inhibitor 2D. Our findings highlight the importance of functional miRNAs in insulinomas.
Collapse
Affiliation(s)
- Xiuli Jiang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases (X.J., A.S., Y.S., Y.C., W.G., W.W., G.N., Y.C.), Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, and Laboratory of Endocrinology and Metabolism (G.N.), Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Aijing Shan
- Shanghai Clinical Center for Endocrine and Metabolic Diseases (X.J., A.S., Y.S., Y.C., W.G., W.W., G.N., Y.C.), Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, and Laboratory of Endocrinology and Metabolism (G.N.), Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Yutong Su
- Shanghai Clinical Center for Endocrine and Metabolic Diseases (X.J., A.S., Y.S., Y.C., W.G., W.W., G.N., Y.C.), Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, and Laboratory of Endocrinology and Metabolism (G.N.), Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Yulong Cheng
- Shanghai Clinical Center for Endocrine and Metabolic Diseases (X.J., A.S., Y.S., Y.C., W.G., W.W., G.N., Y.C.), Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, and Laboratory of Endocrinology and Metabolism (G.N.), Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Weiqiong Gu
- Shanghai Clinical Center for Endocrine and Metabolic Diseases (X.J., A.S., Y.S., Y.C., W.G., W.W., G.N., Y.C.), Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, and Laboratory of Endocrinology and Metabolism (G.N.), Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Weiqing Wang
- Shanghai Clinical Center for Endocrine and Metabolic Diseases (X.J., A.S., Y.S., Y.C., W.G., W.W., G.N., Y.C.), Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, and Laboratory of Endocrinology and Metabolism (G.N.), Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Guang Ning
- Shanghai Clinical Center for Endocrine and Metabolic Diseases (X.J., A.S., Y.S., Y.C., W.G., W.W., G.N., Y.C.), Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, and Laboratory of Endocrinology and Metabolism (G.N.), Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | - Yanan Cao
- Shanghai Clinical Center for Endocrine and Metabolic Diseases (X.J., A.S., Y.S., Y.C., W.G., W.W., G.N., Y.C.), Shanghai Key Laboratory for Endocrine Tumors, Rui-Jin Hospital, Shanghai Jiao-Tong University School of Medicine, and Laboratory of Endocrinology and Metabolism (G.N.), Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
33
|
Srivastava V, Patel B, Kumar M, Shukla M, Pandey M. Cyclin D1, retinoblastoma and p16 protein expression in carcinoma of the gallbladder. Asian Pac J Cancer Prev 2015; 14:2711-5. [PMID: 23803020 DOI: 10.7314/apjcp.2013.14.5.2711] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cancer of the gallbladder is a relatively rare neoplasm with a poor prognosis. The exact mechanisms of its genesis are not known and very little information is available on molecular events leading to labeling this as an orphan cancer. MATERIALS AND METHODS In this prospective case control study we evaluated the expression of p16, pRb and cyclin D1 by immunohistochemistry to study the G1-S cell-cycle check point and its possible role in gallbladder carcinogenesis. A total of 25 patients with gallbladder carcinoma (group I), 25 with cholelithiasis (group II) and 10 normal controls. were enrolled. RESULTS Cyclin D1 expression was seen in 10 (40%) patients each with carcinoma and cholelithiasis while only in 2 (20%) of the normal gallbladders but differences were not statistically significant (p value=0.488). p16 was expressed in 12% patients of carcinoma of the gallbladder and 28% of cholelithiasis, however this difference was not statistically significant (p value=0.095). Retinoblastoma protein was found to be expressed in 50% of normal gallbladders and 6 (24%) of carcinoma and 8 (32%) of gallstones. The present study failed to demonstrate any conclusive role of cyclin D1/RB/ p16 pathway in carcinoma of the gallbladder. CONCLUSIONS The positive relation observed between tumor metastasis and cyclinD1 expression and p16 with nodal metastasis suggested that higher cyclin D1/p16 expression may act as a predictive biomarker for aggressive behavior of gallbladder malignancies.
Collapse
Affiliation(s)
- Vineeta Srivastava
- Department of Surgical Oncology and Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | | | | | | | |
Collapse
|
34
|
Affiliation(s)
- Karen E Sheppard
- Division of Cancer Research; Peter MacCallum Cancer Centre; East Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology; University of Melbourne; Parkville, Victoria, Australia
| | - Richard B Pearson
- Division of Cancer Research; Peter MacCallum Cancer Centre; East Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology; University of Melbourne; Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology; University of Melbourne; Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology; Monash University; Clayton, Victoria, Australia
| | - Ross D Hannan
- Division of Cancer Research; Peter MacCallum Cancer Centre; East Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology; University of Melbourne; Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology; University of Melbourne; Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology; Monash University; Clayton, Victoria, Australia
- School of Biomedical Sciences; University of Queensland; Brisbane, Queensland, Australia
| |
Collapse
|
35
|
P16 INK4A is required for cisplatin resistance in cervical carcinoma SiHa cells. Oncol Lett 2014; 9:1104-1108. [PMID: 25663864 PMCID: PMC4315085 DOI: 10.3892/ol.2014.2814] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 09/26/2014] [Indexed: 01/01/2023] Open
Abstract
Cervical cancer is the third most commonly diagnosed cancer worldwide and the fourth leading cause of cancer-related mortality in females worldwide, accounting for 10-15% of cancer-related mortalities. Cytological screening and DNA testing for high-risk human papillomavirus (HPV) types have markedly decreased the rates of cervical cancer in developed countries, however, for vulnerable populations without access to health care, cervical cancer remains a considerable problem. Chemotherapeutic agents such as cisplatin (DDP) are considered as first-line treatment for cervical carcinoma. Although initially patients often exhibit high responsiveness, the majority eventually develop DDP resistance. However, the mechanisms underlying this process remain unclear. Furthermore, patients with metastatic cancer and those exhibiting persistent or recurrent disease after platinum-based chemoradiotherapy have limited options and thus, non-platinum combination chemotherapy has been proposed as a strategy to circumvent platinum resistance, however, novel therapeutic strategies are required. In the present study, P16 expression was analyzed by quantitative-polymerase chain reaction and western blot analysis in SiHa and SiHa-DDP cells and the interaction between P16 and CDK4 was detected via co-immunoprecipitation. In addition, the proliferation and apoptosis rates of P16 knockdown SiHa-DDP cells were measured by MTT assay and Annexin V flow cytometry and the subsequent changes in cyclin D1 and pRb expression were analyzed by western blot analysis. In this study, a high level of P16INK4A expression and its enhanced interaction with cyclin-dependent kinase-4 in cervical carcinoma DDP-resistance cells (SiHa-DDP) was identified, which was associated with the inactivation of phosphorylated retinoblastoma protein (pRb). Knockdown of P16INK4A significantly induced cellular growth, when compared with the control cells, via the upregulation of pRb, and also promoted apoptosis following treatment with DDP. The results of this study indicated, for the first time, that P16INK4A is required for DDP resistance in cervical carcinoma SiHa cells and, thus, these results may lead to the development of novel strategies for the treatment of chemoresistant cervical carcinoma.
Collapse
|
36
|
Sonzogni SV, Ogara MF, Castillo DS, Sirkin PF, Radicella JP, Cánepa ET. Nuclear translocation of p19INK4d in response to oxidative DNA damage promotes chromatin relaxation. Mol Cell Biochem 2014; 398:63-72. [DOI: 10.1007/s11010-014-2205-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 08/30/2014] [Indexed: 12/23/2022]
|
37
|
Fucoidan inhibits the proliferation of human urinary bladder cancer T24 cells by blocking cell cycle progression and inducing apoptosis. Molecules 2014; 19:5981-98. [PMID: 24818577 PMCID: PMC6271230 DOI: 10.3390/molecules19055981] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 11/16/2022] Open
Abstract
Although fucoidan has been shown to exert anticancer activity against several types of cancer cell lines, no reports have explored fucoidan-affected cell growth in human urinary bladder cancer cells. In this study, we investigated the anti-proliferative effects of fucoidan in human bladder cancer T24 cells. Our results indicated that fucoidan decreased the viability of T24 cells through the induction of G1 arrest and apoptosis. Fucoidan-induced G1 arrest is associated with the enhanced expression of the Cdk inhibitor p21WAF1/CIP1 and dephosphorylation of the pRB along with enhanced binding of p21 to Cdk4/6 as well as pRB to the transcription factor E2Fs. Further investigations showed the loss of mitochondrial membrane potential and the release of cytochrome c from mitochondria to cytosol, proving mitochondrial dysfunction upon fucoidan treatment with a corresponding increase in the Bax/Bcl-2 expression ratio. Fucoidan-triggered apoptosis was also accompanied by the up-regulation of Fas and truncated Bid as well as the sequential activation of caspase-8. Furthermore, a significant increased activation of caspase-9/-3 was detected in response to fucoidan treatment with the decreased expression of IAPs and degradation of PARP, whereas a pan-caspase inhibitor significantly suppressed apoptosis and rescued the cell viability reduction. In conclusion, these observations suggest that fucoidan attenuates G1-S phase cell cycle progression and serves as an important mediator of crosstalk between caspase-dependent intrinsic and extrinsic apoptotic pathways in T24 cells.
Collapse
|
38
|
p19Ink4d is a tumor suppressor and controls pituitary anterior lobe cell proliferation. Mol Cell Biol 2014; 34:2121-34. [PMID: 24687853 DOI: 10.1128/mcb.01363-13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Pituitary tumors develop in about one-quarter of the population, and most arise from the anterior lobe (AL). The pituitary gland is particularly sensitive to genetic alteration of genes involved in the cyclin-dependent kinase (CDK) inhibitor (CKI)-CDK-retinoblastoma protein (Rb) pathway. Mice heterozygous for the Rb mutation develop pituitary tumors, with about 20% arising from the AL. Perplexingly, none of the CKI-deficient mice reported thus far develop pituitary AL tumors. In this study, we show that deletion of p19(Ink4d) (p19), a CKI gene, in mice results in spontaneous development of tumors in multiple organs and tissues. Specifically, more than one-half of the mutant mice developed pituitary hyperplasia or tumors predominantly in the AL. Tumor development is associated with increased cell proliferation and enhanced activity of Cdk4 and Cdk6 and phosphorylation of Rb protein. Though Cdk4 is indispensable for postnatal pituitary cell proliferation, it is not required for the hyperproliferative pituitary phenotype caused by p19 loss. Loss of p19 phosphorylates Rb in Cdk4(-/-) pituitary AL cells and mouse embryonic fibroblasts (MEFs) and rescues their proliferation defects, at least partially, through the activation of Cdk6. These results provide the first genetic evidence that p19 is a tumor suppressor and the major CKI gene that controls pituitary AL cell proliferation.
Collapse
|
39
|
Liang X, Wang P, Gao Q, Tao X. Exogenous activation of LKB1/AMPK signaling induces G₁ arrest in cells with endogenous LKB1 expression. Mol Med Rep 2014; 9:1019-24. [PMID: 24469340 DOI: 10.3892/mmr.2014.1916] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 01/17/2014] [Indexed: 11/06/2022] Open
Abstract
The tumor suppressor protein LKB1 is a serine/threonine kinase that plays a critical role in cell proliferation, and its inactivation has been linked to tumorigenesis in various cancer types. Current understanding of the LKB1 function is largely restricted to results from experiments on LKB1‑deficient cancer cells, while the regulation and activity of endogenous LKB1 has been rarely investigated. In a previous study, we showed that LKB1 knockdown in two healthy cell lines accelerates cell cycle progression through the G1/S checkpoint by inhibition of the p53 and p16 pathways. In the present study, we examined the effects of overexpression of LKB1 on two healthy and one cancer cell line. Administration of exogenous LKB1 activated LKB1/AMPK signaling and arrested the cell cycle at the G1 phase in an LKB1-dependent manner. G1 arrest induced by LKB1 was accompanied by the downregulation of cyclin D1 and cyclin D3, and the upregulation of p53, p21 and p16, while no differences were detected for CDK4, CDK6, cyclin E, p15 and p27. These results indicated that exogenous activation of LKB1/AMPK signaling inhibits the G1/S cell cycle transition, even in cells with an endogenous expression of LKB1. Findings of the present study extend earlier observations on LKB1‑inactivated neoplastic cells and provide novel insights into the growth-inhibitory effects of LKB1.
Collapse
Affiliation(s)
- Xiaoyan Liang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Pilong Wang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Qing Gao
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiaohong Tao
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
40
|
Kreis NN, Sanhaji M, Rieger MA, Louwen F, Yuan J. p21Waf1/Cip1 deficiency causes multiple mitotic defects in tumor cells. Oncogene 2013; 33:5716-28. [PMID: 24317508 DOI: 10.1038/onc.2013.518] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 10/23/2013] [Accepted: 10/25/2013] [Indexed: 12/12/2022]
Abstract
As a multifaceted molecule, p21 plays multiple critical roles in cell cycle regulation, differentiation, apoptosis, DNA repair, senescence, aging and stem cell reprogramming. The important roles of p21 in the interphase of the cell cycle have been intensively investigated. The function of p21 in mitosis has been proposed but not systematically studied. We show here that p21 is abundant in mitosis and binds to and inhibits the activity of Cdk1/cyclin B1. Deficiency of p21 prolongs the duration of mitosis by extending metaphase, anaphase and cytokinesis. The activity of Aurora B is reduced and the localization of Aurora B on the central spindle is disturbed in anaphase cells without p21. Moreover, HCT116 p21-/-, HeLa and Saos-2 cells depleted of p21 encounter problems in chromosome segregation and cytokinesis. Gently inhibiting the mitotic Cdk1 or add-back of p21 rescues segregation defect in HCT116 p21-/- cells. Our data demonstrate that p21 is important for a fine-tuned control of the Cdk1 activity in mitosis, and its proper function facilitates a smooth mitotic progression. Given that p21 is downregulated in the majority of tumors, either by the loss of tumor suppressors like p53 or by hyperactive oncogenes such as c-myc, this finding also sheds new light on the molecular mechanisms by which p21 functions as a tumor suppressor.
Collapse
Affiliation(s)
- N-N Kreis
- Department of Gynecology and Obstetrics, Frankfurt, Germany
| | - M Sanhaji
- Department of Gynecology and Obstetrics, Frankfurt, Germany
| | - M A Rieger
- 1] Department of Hematology/Oncology, J W Goethe-University, Theodor-Stern-Kai 7, Frankfurt, Germany [2] Georg-Speyer-Haus, Frankfurt, Germany [3] German Cancer Consortium (DKTK), Heidelberg, Germany [4] German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - F Louwen
- Department of Gynecology and Obstetrics, Frankfurt, Germany
| | - J Yuan
- Department of Gynecology and Obstetrics, Frankfurt, Germany
| |
Collapse
|
41
|
Abstract
PURPOSE To investigate the novel application of tissue microarray (TMA) technology to corneal disease and to report altered protein expression of senescence-associated cyclin-dependent kinase inhibitors p21 and p16 in Fuchs endothelial corneal dystrophy (FECD). METHODS A TMA including 208 cores was generated from paraffin-embedded tissues, including corneal buttons of 50 FECD and 5 keratoconus patients retrieved after penetrating keratoplasty, 10 autopsy globes with nonpathologic corneas, and nonocular control specimens. TMA sections were immunolabeled for p21 and p16 and analyzed using a 9-grade scoring system (0-8). Result validation was performed by immunolabeling of individual whole tissue sections. Corneal endothelial p21 and p16 expression levels in FECD specimens compared with controls served as main outcome measures. RESULTS TMA immunohistochemical analysis disclosed increased endothelial expression levels of nuclear p21 in FECD specimens (P < 0.05) and an altered endothelial p16 expression pattern. Immunolabeling of whole tissue sections showed statistically significant endothelial overexpression of both proteins (p21 and p16, P < 0.05). CONCLUSIONS The present study introduces TMA technology as a valuable tool for molecular high-throughput profiling of corneal tissues. It demonstrates p21 and p16 overexpression in the corneal endothelium of genetically undifferentiated FECD patients supporting a role of cellular senescence in the pathogenesis of FECD.
Collapse
|
42
|
Al-Khalaf HH, Aboussekhra A. p16(INK4A) positively regulates p21(WAF1) expression by suppressing AUF1-dependent mRNA decay. PLoS One 2013; 8:e70133. [PMID: 23894605 PMCID: PMC3720951 DOI: 10.1371/journal.pone.0070133] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Accepted: 06/17/2013] [Indexed: 01/01/2023] Open
Abstract
Background p16INK4a and p21WAF1 are two independent cyclin-dependent kinase inhibitors encoded by the CDKN2A and CDKN1A genes, respectively. p16INK4a and p21WAF1 are similarly involved in various anti-cancer processes, including the regulation of the critical G1 to S phase transition of the cell cycle, senescence and apoptosis. Therefore, we sought to elucidate the molecular mechanisms underlying the link between these two important tumor suppressor proteins. Methodology/Principal Findings We have shown here that the p16INK4a protein positively controls the expression of p21WAF1 in both human and mouse cells. p16INK4a stabilizes the CDKN1A mRNA through negative regulation of the mRNA decay-promoting AUF1 protein. Immunoprecipitation of AUF1-associated RNAs followed by quantitative RT-PCR indicated that endogenous AUF1 binds to the CDKN1A mRNA in a p16INK4A-dependent manner. Furthermore, while AUF1 down-regulation increased the expression level of the CDKN1A mRNA, the concurrent knockdown of AUF1 and CDKN2A, using specific silencing RNAs, restored the normal expression of the gene. Moreover, we used EGFP reporter fused to the CDKN2A AU-rich element (ARE) to demonstrate that p16INK4A regulation of the CDKN1A mRNA is AUF1- and ARE-dependent. Furthermore, ectopic expression of p16INK4A in p16INK4A-deficient breast epithelial MCF-10A cells significantly increased the level of p21WAF1, with no effect on cell proliferation. In addition, we have shown direct correlation between p16INK4a and p21WAF1 levels in various cancer cell lines. Conclusion/Significance These findings show that p16INK4a stabilizes the CDKN1A mRNA in an AUF1-dependent manner, and further confirm the presence of a direct link between the 2 important cancer-related pathways, pRB/p16INK4A and p14ARF/p53/p21WAF1.
Collapse
Affiliation(s)
- Huda H. Al-Khalaf
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- The Joint Center for Genomics Research, King Abdulaziz City for Science and Technology, Riyadh, KSA
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- * E-mail:
| |
Collapse
|
43
|
Al-Khalaf HH, Aboussekhra A. ATR controls the UV-related upregulation of the CDKN1A mRNA in a Cdk1/HuR-dependent manner. Mol Carcinog 2013; 53:979-87. [PMID: 23813879 DOI: 10.1002/mc.22066] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2013] [Revised: 05/13/2013] [Accepted: 05/31/2013] [Indexed: 11/10/2022]
Abstract
Ultraviolet (UV) light is a carcinogenic agent that upregulates the expression of several genes involved in various cellular processes, including cell cycle checkpoints and apoptosis. The universal cyclin-dependent kinase inhibitor p21(WAF1/Cip1) plays major roles in these processes, and the level of its corresponding message increases several times in response to UV-induced DNA damage. This upregulation is mainly posttranscriptional owing to HuR-dependent mRNA stabilization. Since the protein kinase Atr plays major roles during the cellular response to UV damage, we sought to investigate its possible implication in the stabilization of the p21(WAF1/Cip1) coding mRNA. We have shown that the UV-dependent accumulation of the CDKN1A mRNA is indeed under the control of the Atr protein kinase. Upon UV damage, Atr allows nuclear-cytoplasmic shuttling of the HuR protein, which binds the CDKN1A mRNA and reduces its turnover. This ATR-dependent effect is mediated through UV-related phosphorylation/inactivation of the Cdk1 protein kinase by Atr, which leads to the dissociation of HuR from Cdk1. Indeed, inhibition or shRNA specific knockdown of CDK1 in ATR-deficient cells enhanced the cytoplasmic level of HuR and restored the CDKN1A mRNA upregulation in response to UV damage. These results show that ATR stabilizes the CDKN1A message in response to UV damage through Cdk1-related cytoplasmic accumulation of HuR.
Collapse
Affiliation(s)
- Huda H Al-Khalaf
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Center, Riyadh, Kingdom of Saudi Arabia; The Joint Center for Genomics Research, King Abdulaziz City for Science and Technology, Riyadh, Kingdom of Saudi Arabia
| | | |
Collapse
|
44
|
Bisteau X, Paternot S, Colleoni B, Ecker K, Coulonval K, De Groote P, Declercq W, Hengst L, Roger PP. CDK4 T172 phosphorylation is central in a CDK7-dependent bidirectional CDK4/CDK2 interplay mediated by p21 phosphorylation at the restriction point. PLoS Genet 2013; 9:e1003546. [PMID: 23737759 PMCID: PMC3667761 DOI: 10.1371/journal.pgen.1003546] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 04/22/2013] [Indexed: 01/24/2023] Open
Abstract
Cell cycle progression, including genome duplication, is orchestrated by cyclin-dependent kinases (CDKs). CDK activation depends on phosphorylation of their T-loop by a CDK–activating kinase (CAK). In animals, the only known CAK for CDK2 and CDK1 is cyclin H-CDK7, which is constitutively active. Therefore, the critical activation step is dephosphorylation of inhibitory sites by Cdc25 phosphatases rather than unrestricted T-loop phosphorylation. Homologous CDK4 and CDK6 bound to cyclins D are master integrators of mitogenic/oncogenic signaling cascades by initiating the inactivation of the central oncosuppressor pRb and cell cycle commitment at the restriction point. Unlike the situation in CDK1 and CDK2 cyclin complexes, and in contrast to the weak but constitutive T177 phosphorylation of CDK6, we have identified the T-loop phosphorylation at T172 as the highly regulated step determining CDK4 activity. Whether both CDK4 and CDK6 phosphorylations are catalyzed by CDK7 remains unclear. To answer this question, we took a chemical-genetics approach by using analogue-sensitive CDK7(as/as) mutant HCT116 cells, in which CDK7 can be specifically inhibited by bulky adenine analogs. Intriguingly, CDK7 inhibition prevented activating phosphorylations of CDK4/6, but for CDK4 this was at least partly dependent on its binding to p21cip1. In response to CDK7 inhibition, p21-binding to CDK4 increased concomitantly with disappearance of the most abundant phosphorylation of p21, which we localized at S130 and found to be catalyzed by both CDK4 and CDK2. The S130A mutation of p21 prevented the activating CDK4 phosphorylation, and inhibition of CDK4/6 and CDK2 impaired phosphorylations of both p21 and p21-bound CDK4. Therefore, specific CDK7 inhibition revealed the following: a crucial but partly indirect CDK7 involvement in phosphorylation/activation of CDK4 and CDK6; existence of CDK4-activating kinase(s) other than CDK7; and novel CDK7-dependent positive feedbacks mediated by p21 phosphorylation by CDK4 and CDK2 to sustain CDK4 activation, pRb inactivation, and restriction point passage. In the cell cycle, duplication of all the cellular components and subsequent cell division are governed by a family of protein kinases associated with cyclins (CDKs). Related CDK4 and CDK6 bound to cyclins D are the first CDKs to be activated in response to cell proliferation signals. They thus play a central role in the cell multiplication decision, especially in most cancer cells in which CDK4 activity is highly deregulated. We have identified the activating T172 phosphorylation instead of cyclin D expression as the highly regulated step determining CDK4 activation. This finding contrasts with the prevalent view that the only identified metazoan CDK-activating kinase, CDK7, is constitutively active. By using human cells genetically engineered for specific chemical inhibition of CDK7, we found that CDK7 activity was indeed required for CDK4 activation. However, this dependence was conditioned by CDK4 binding to the CDK inhibitory protein p21, which increased in response to CDK7 inhibition. Further investigation revealed that CDK7 inhibition affects a major phosphorylation of p21, which we found to be required for CDK4 activation and performed by CDK4 itself and CDK2. Thus, depending on CDK7 activity, CDK4 and CDK2 facilitate CDK4 activation, generating novel positive feedbacks involved in the cell cycle decision.
Collapse
Affiliation(s)
- Xavier Bisteau
- WELBIO and Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Sabine Paternot
- WELBIO and Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Bianca Colleoni
- WELBIO and Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Karin Ecker
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Katia Coulonval
- WELBIO and Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Philippe De Groote
- Department for Molecular Biomedical Research, VIB, and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wim Declercq
- Department for Molecular Biomedical Research, VIB, and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ludger Hengst
- Division of Medical Biochemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Pierre P. Roger
- WELBIO and Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
- * E-mail:
| |
Collapse
|
45
|
Kim KN, Ahn G, Heo SJ, Kang SM, Kang MC, Yang HM, Kim D, Roh SW, Kim SK, Jeon BT, Park PJ, Jung WK, Jeon YJ. Inhibition of tumor growth in vitro and in vivo by fucoxanthin against melanoma B16F10 cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2013; 35:39-46. [PMID: 23228706 DOI: 10.1016/j.etap.2012.10.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 10/02/2012] [Accepted: 10/05/2012] [Indexed: 05/24/2023]
Abstract
The present study was designed to evaluate the molecular mechanisms of fucoxanthin against melanoma cell lines (B16F10 cells). Fucoxanthin reduced the proliferation of B16F10 cells in a dose-dependent manner accompanied by the induction of cell cycle arrest during the G(0)/G(1) phase and apoptosis. Fucoxanthin-induced G(0)/G(1) arrest was associated with a marked decrease in the protein expressions of phosphorylated-Rb (retinoblastoma protein), cyclin D (1 and 2) and cyclin-dependent kinase (CDK) 4 and up-regulation of the protein levels of p15(INK4B) and p27(Kip1). Fucoxanthin-induced apoptosis was accompanied with the down-regulation of the protein levels of Bcl-xL, an inhibitor of apoptosis proteins (IAPs), resulting in a sequential activation of caspase-9, caspase-3, and PARP. Furthermore, the anti-tumor effect of fucoxanthin was assessed in vivo in Balb/c mice. Intraperitoneal administration of fucoxanthin significantly inhibited the growth of tumor mass in B16F10 cells implanted mice.
Collapse
Affiliation(s)
- Kil-Nam Kim
- Marine Bio Research Team, Korea Basic Science Institute (KBSI), Jeju 690-140, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Ni HJ, Chang YN, Kao PH, Chai SP, Hsieh YH, Wang DH, Fong JC. Depletion of SUMO ligase hMMS21 impairs G1 to S transition in MCF-7 breast cancer cells. Biochim Biophys Acta Gen Subj 2012; 1820:1893-900. [DOI: 10.1016/j.bbagen.2012.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 07/20/2012] [Accepted: 08/06/2012] [Indexed: 10/28/2022]
|
47
|
Bai F, Smith MD, Chan HL, Pei XH. Germline mutation of Brca1 alters the fate of mammary luminal cells and causes luminal-to-basal mammary tumor transformation. Oncogene 2012; 32:2715-25. [PMID: 22777348 DOI: 10.1038/onc.2012.293] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Breast cancer developed in familial BRCA1 mutation carriers bears striking similarities to sporadic basal-like breast tumors. The mechanism underlying the function of BRCA1 in suppressing basal-like breast cancer remains unclear. We previously reported that the deletion of p18(Ink4c) (p18), an inhibitor of G1 cyclin Ds-dependent CDK4 and CDK6, stimulates mammary luminal progenitor cell proliferation and leads to spontaneous luminal tumor development. We report here that germline mutation of Brca1 in p18-deficient mice blocks the increase of luminal progenitor cells, impairs luminal gene expression and promotes malignant transformation of mammary tumors. Instead of the luminal mammary tumors developed in p18 single-mutant mice, mammary tumors developed in the p18;Brca1 mice, similar to breast cancer developed in familial BRCA1 carriers, exhibited extensive basal-like features and lost the remaining wild-type allele of Brca1. These results reveal distinct functions of the RB and BRCA1 pathways in suppressing luminal and basal-like mammary tumors, respectively. These results also suggest a novel mechanism--causing luminal-to-basal transformation--for the development of basal-like breast cancer in familial BRCA1 carriers and establish a unique mouse model for developing therapeutic strategies to target both luminal and basal-like breast cancers.
Collapse
Affiliation(s)
- F Bai
- Molecular Oncology Program, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | |
Collapse
|
48
|
CDK2 and PKA mediated-sequential phosphorylation is critical for p19INK4d function in the DNA damage response. PLoS One 2012; 7:e35638. [PMID: 22558186 PMCID: PMC3338453 DOI: 10.1371/journal.pone.0035638] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 03/19/2012] [Indexed: 12/20/2022] Open
Abstract
DNA damage triggers a phosphorylation-based signaling cascade known as the DNA damage response. p19INK4d, a member of the INK4 family of CDK4/6 inhibitors, has been reported to participate in the DNA damage response promoting DNA repair and cell survival. Here, we provide mechanistic insight into the activation mechanism of p19INK4d linked to the response to DNA damage. Results showed that p19INK4d becomes phosphorylated following UV radiation, β-amyloid peptide and cisplatin treatments. ATM-Chk2/ATR-Chk1 signaling pathways were found to be differentially involved in p19INK4d phosphorylation depending on the type of DNA damage. Two sequential phosphorylation events at serine 76 and threonine 141 were identified using p19INK4d single-point mutants in metabolic labeling assays with 32P-orthophosphate. CDK2 and PKA were found to participate in p19INK4d phosphorylation process and that they would mediate serine 76 and threonine 141 modifications respectively. Nuclear translocation of p19INK4d induced by DNA damage was shown to be dependent on serine 76 phosphorylation. Most importantly, both phosphorylation sites were found to be crucial for p19INK4d function in DNA repair and cell survival. In contrast, serine 76 and threonine 141 were dispensable for CDK4/6 inhibition highlighting the independence of p19INK4d functions, in agreement with our previous findings. These results constitute the first description of the activation mechanism of p19INK4d in response to genotoxic stress and demonstrate the functional relevance of this activation following DNA damage.
Collapse
|
49
|
Zhang Y, Yan B. Cell cycle regulation by carboxylated multiwalled carbon nanotubes through p53-independent induction of p21 under the control of the BMP signaling pathway. Chem Res Toxicol 2012; 25:1212-21. [PMID: 22428663 DOI: 10.1021/tx300059m] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This report describes how carboxylated multiwalled carbon nanotubes (MWCNTs) induce p53-independent p21 expression and cell cycle arrest. MWCNTs suppress BMP signaling and lead to the downregulation of Id protein production and the upregulation of p21 because p21 expression is directly controlled by Id proteins through their regulation of the E-box motifs in the p21 promoter. The overexpressed p21 protein then binds to the cyclin D/cdk4,6 complexes and inhibits the phosphorylation of Rb protein. Hypophosphorylation of Rb prevents the release of E2F factors and causes cell cycle arrest. These findings provide valuable insight into a mechanistic understanding of carbon nanotubes' effects on cellular functions.
Collapse
Affiliation(s)
- Yi Zhang
- School of Pharmaceutical Sciences, Shandong University, Jinan 250100, China
| | | |
Collapse
|
50
|
Taccaliti A, Silvetti F, Palmonella G, Boscaro M. Anaplastic thyroid carcinoma. Front Endocrinol (Lausanne) 2012; 3:84. [PMID: 22783225 PMCID: PMC3389605 DOI: 10.3389/fendo.2012.00084] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 06/15/2012] [Indexed: 02/01/2023] Open
Abstract
Thyroid cancers represent about 1% of all human cancers. Differentiate thyroid carcinomas (DTCs), papillary and follicular cancers, are the most frequent forms, instead Anaplastic Thyroid Carcinoma (ATC) is estimated to comprise 1-2% of thyroid malignancies and it accounts for 14-39% of thyroid cancer deaths. The annual incidence of ATC is about one to two cases/million, with the overall incidence being higher in Europe (and area of endemic goiter) than in USA. ATC has a more complex genotype than DTCs, with chromosomal aberrations present in 85-100% of cases. A small number of gene mutations have been identified, and there appears to be a progression in mutations acquired during dedifferentiation. The mean survival time is around 6 months from diagnosis an outcome that is frequently not altered by treatment. ATC presents with a rapidly growing fixed and hard neck mass, often metastatic local lymph nodes appreciable on examination and/or vocal paralysis. Symptoms may reflect rapid growth of tumor with local invasion and/or compression. The majority of patients with ATC die from aggressive local regional disease, primarily from upper airway respiratory failure. For this reason, aggressive local therapy is indicated in all patients who can tolerate it. Although rarely possible, complete surgical resection gives the best chance of long-term control and improved survival. Therapy options include surgery, external beam radiation therapy, tracheostomy, chemotherapy, and investigational clinical trials. Multimodal or combination therapy should be useful. In fact, surgical debulking of local tumor, combined with external beam radiation therapy and chemotherapy as neoadjuvant (before surgery) or adjuvant (after surgery) therapy, may prevent death from local airway obstruction and as best may slight prolong survival. Investigational clinical trials in phase I or in phase II are actually in running and they include anti-angiogenetic drugs, multi-kinase inhibitor drugs.
Collapse
Affiliation(s)
- Augusto Taccaliti
- Division of Endocrinology, Azienda Ospedaliero Universitaria Torrette – AnconaAncona, Italy
- *Correspondence: Augusto Taccaliti, Division of Endocrinology, Azienda Ospedaliero Universitaria Torrette – Ancona, Via Conca 71, 60126 Ancona, Italy. e-mail:
| | - Francesca Silvetti
- Division of Endocrinology, Azienda Ospedaliero Universitaria Torrette – AnconaAncona, Italy
| | - Gioia Palmonella
- Division of Endocrinology, Azienda Ospedaliero Universitaria Torrette – AnconaAncona, Italy
| | - Marco Boscaro
- Division of Endocrinology, Azienda Ospedaliero Universitaria Torrette – AnconaAncona, Italy
| |
Collapse
|