1
|
Barbeiro CDO, Fernandes D, Palaçon MP, Castilho RM, de Almeida LY, Bufalino A. Inflammatory Cells Can Alter the Levels of H3K9ac and γH2AX in Dysplastic Cells and Favor Tumor Phenotype. J Pers Med 2023; 13:jpm13040662. [PMID: 37109048 PMCID: PMC10141380 DOI: 10.3390/jpm13040662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/06/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Oral potentially malignant disorders (OPMD) are clinical presentations that carry an increased risk of cancer development. Currently, epithelial dysplasia grade is based on architectural and cytological epithelial changes and is used to predict the malignant transformation of these lesions. However, predicting which OPMD will progress to a malignant tumor is very challenging. Inflammatory infiltrates can favor cancer development, and recent studies suggest that this association with OPMD lesions may be related to the etiology and/or aggressive clinical behavior of these lesions. Epigenetic changes such as histone modifications may mediate chronic inflammation and also favor tumor cells in immune resistance and evasion. This study aimed to evaluate the relationship between histone acetylation (H3K9ac) and DNA damage in the context of dysplastic lesions with prominent chronic inflammation. Immunofluorescence of "low-risk" and "high-risk" OPMD lesions (n = 24) and inflammatory fibrous hyperplasia (n = 10) as the control group was performed to assess histone acetylation levels and DNA damage through the phosphorylation of H2AX (γH2AX). Cell co-culture assays with PBMCs and oral keratinocyte cell lines (NOK-SI, DOK, and SCC-25) were performed to assess proliferation, adhesion, migration, and epithelial-mesenchymal transition (EMT). Oral dysplastic lesions showed a hypoacetylation of H3K9 and low levels of γH2AX compared to control. The contact of dysplastic oral keratinocytes with PBMCs favored EMT and the loss of cell-cell adhesion. On the other hand, p27 levels increased and cyclin E decreased in DOK, indicating cell cycle arrest. We conclude that the presence of chronic inflammation associated to dysplastic lesions is capable of promoting epigenetic alterations, which in turn can favor the process of malignant transformation.
Collapse
Affiliation(s)
- Camila de Oliveira Barbeiro
- Oral Medicine, Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (Unesp), Araraquara 14801-903, SP, Brazil
| | - Darcy Fernandes
- Oral Medicine, Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (Unesp), Araraquara 14801-903, SP, Brazil
| | - Mariana Paravani Palaçon
- Oral Medicine, Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (Unesp), Araraquara 14801-903, SP, Brazil
| | - Rogerio Moraes Castilho
- Laboratory of Epithelial Biology, Department of Periodontics and Oral Medicine, University of Michigan, 1011N University Av, Ann Arbor, MI 48109-1078, USA
| | - Luciana Yamamoto de Almeida
- Oral Medicine, Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (Unesp), Araraquara 14801-903, SP, Brazil
| | - Andreia Bufalino
- Oral Medicine, Department of Diagnosis and Surgery, School of Dentistry, São Paulo State University (Unesp), Araraquara 14801-903, SP, Brazil
| |
Collapse
|
2
|
Expression of cyclin D1 correlates with p27KIP1 and regulates the degree of oral dysplasia and squamous cell carcinoma differentiation. Oral Surg Oral Med Oral Pathol Oral Radiol 2018; 126:174-183. [DOI: 10.1016/j.oooo.2018.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 01/16/2018] [Accepted: 01/23/2018] [Indexed: 01/30/2023]
|
3
|
Clinicopathological and Prognostic Significance of p27 Expression in Oral Squamous Cell Carcinoma: A Meta-Analysis. Int J Biol Markers 2018; 28:e329-35. [PMID: 23787492 DOI: 10.5301/jbm.5000035] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2013] [Indexed: 12/18/2022]
Abstract
Despite being already known that p27 can regulate cell proliferation, cell motility and apoptosis, the role of p27 expression in oral squamous cell carcinoma (OSCC) remains controversial. The purpose of this study was to comprehensively evaluate, with a meta-analysis, the clinicopathological and prognostic role of p27 expression in OSCC. A meta-analysis of eligible studies was performed to assess the effects of p27 expression on clinicopathological parameters and overall survival (OS) in patients with OSCC, using pooled relative risks with 95% confidence intervals. Heterogeneity and publication bias were also assessed. Fourteen studies involving a total of 1,010 patients met the inclusion criteria. Low p27 expression was significantly associated with advanced TNM stage (p<0.001), worse histology (p=0.025), and lymph node metastasis (p<0.001), but not with tumor size (p=0.181). The pooled RR of 0.743 (p=0.002) suggested that low p27 expression has a poor prognosis in patients with OSCC. A significant heterogeneity among studies was detected for lymph node status (χ2=34.60, I2=68.2%, p<0.001) and OS (χ2=14.86, I2=39.4%, p=0.095). We did not detect a significant publication bias in this meta-analysis. Our meta-analysis suggests that p27 expression status might be useful as a predictive biomarker in clinical practice, and might potently predict OS in OSCC patients.
Collapse
|
4
|
Danforth DN. Genomic Changes in Normal Breast Tissue in Women at Normal Risk or at High Risk for Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2016; 10:109-46. [PMID: 27559297 PMCID: PMC4990153 DOI: 10.4137/bcbcr.s39384] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/17/2016] [Accepted: 04/19/2016] [Indexed: 12/12/2022]
Abstract
Sporadic breast cancer develops through the accumulation of molecular abnormalities in normal breast tissue, resulting from exposure to estrogens and other carcinogens beginning at adolescence and continuing throughout life. These molecular changes may take a variety of forms, including numerical and structural chromosomal abnormalities, epigenetic changes, and gene expression alterations. To characterize these abnormalities, a review of the literature has been conducted to define the molecular changes in each of the above major genomic categories in normal breast tissue considered to be either at normal risk or at high risk for sporadic breast cancer. This review indicates that normal risk breast tissues (such as reduction mammoplasty) contain evidence of early breast carcinogenesis including loss of heterozygosity, DNA methylation of tumor suppressor and other genes, and telomere shortening. In normal tissues at high risk for breast cancer (such as normal breast tissue adjacent to breast cancer or the contralateral breast), these changes persist, and are increased and accompanied by aneuploidy, increased genomic instability, a wide range of gene expression differences, development of large cancerized fields, and increased proliferation. These changes are consistent with early and long-standing exposure to carcinogens, especially estrogens. A model for the breast carcinogenic pathway in normal risk and high-risk breast tissues is proposed. These findings should clarify our understanding of breast carcinogenesis in normal breast tissue and promote development of improved methods for risk assessment and breast cancer prevention in women.
Collapse
Affiliation(s)
- David N Danforth
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Zhao Y, Li Q, Wu X, Chen P. Upregulation of p27Kip1 by demethylation sensitizes cisplatin-resistant human ovarian cancer SKOV3 cells. Mol Med Rep 2016; 14:1659-66. [PMID: 27314502 DOI: 10.3892/mmr.2016.5399] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/29/2016] [Indexed: 11/06/2022] Open
Abstract
Ovarian cancer has a poor prognosis due to its chemoresistance, and p27Kip1 (p27) has been implicated in tumor prognosis and drug-resistance. However, the regulatory mechanisms of p27 in drug‑resistance in ovarian cancer remain unknown. The current study successfully established chemoresistant cell lines using paclitaxel (TAX), cisplatin (DDP) and carboplatin (CBP) in SKOV3 ovarian cancer cells. The results indicated that the expression levels of p27 were dramatically downregulated in chemoresistant cells. However, 5-aza-2'-deoxycytidine (5-aza) treatment restored p27 expression in DDP-resistant cells, and increased their sensitivity to DDP. In addition, it was observed that the methylation of DDP‑resistant cells, which was downregulated by 5‑aza treatment, was significantly higher compared with SKOV3 cells. Additionally, the overexpression of p27 arrested the cell cycle in S phase and promoted an apoptotic response to DDP. In conclusion, p27 was involved in chemoresistance of SKOV3 cells. Upregulated p27 expression induced by demethylation may enhance sensitivity to DDP through the regulation of the cell cycle.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Gynecology and Obstetrics, The Maternal and Child Health Hospital of Hunan, Changsha, Hunan 410008, P.R. China
| | - Qiaoyan Li
- Department of Gynecology and Obstetrics, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Xiaoying Wu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Puxiang Chen
- Department of Gynecology and Obstetrics, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
6
|
Haque I, Banerjee S, De A, Maity G, Sarkar S, Majumdar M, Jha SS, McGragor D, Banerjee SK. CCN5/WISP-2 promotes growth arrest of triple-negative breast cancer cells through accumulation and trafficking of p27(Kip1) via Skp2 and FOXO3a regulation. Oncogene 2014; 34:3152-63. [PMID: 25132260 DOI: 10.1038/onc.2014.250] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Revised: 04/18/2014] [Accepted: 07/03/2014] [Indexed: 02/07/2023]
Abstract
The matricellular protein CCN5/WISP-2 represents a promising target in triple-negative breast cancer (TNBC) because treatment or induced activation of CCN5 in TNBC cells promotes cell growth arrest at the G0/G1 phase, reduces cell proliferation and delays tumor growth in the xenograft model. Our studies found that the p27(Kip1) tumor suppressor protein is upregulated and relocalized to the nucleus from cytoplasm by CCN5 in these cells and that these two events (upregulation and relocalization of p27(Kip1)) are critical for CCN5-induced growth inhibition of TNBC cells. In the absence of CCN5, p27(Kip1) resides mostly in the cytoplasm, which is associated with the aggressive nature of cancer cells. Mechanistically, CCN5 inhibits Skp2 expression, which seems to stabilize the p27(Kip1) protein in these cells. On the other hand, CCN5 also recruits FOXO3a to mediate the transcriptional regulation of p27(Kip1). The recruitment of FOXO3a is achieved by the induction of its expression and activity through shifting from cytoplasm to the nucleus. Our data indicate that CCN5 blocks PI3K/AKT signaling to dephosphorylate at S318, S253 and Thr32 in FOXO3a for nuclear relocalization and activation of FOXO3a. Moreover, inhibition of α6β1 receptors diminishes CCN5 action on p27(Kip1) in TNBC cells. Collectively, these data suggest that CCN5 effectively inhibits TNBC growth through the accumulation and trafficking of p27(Kip1) via Skp2 and FOXO3a regulation, and thus, activation of CCN5 may have the therapeutic potential to kill TNBC.
Collapse
Affiliation(s)
- I Haque
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - S Banerjee
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - A De
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - G Maity
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | - S Sarkar
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Department of Anatomy and Cell Biology and Department of Pathology, University of Kansas Medical Center, Kansas City, MO, USA
| | - M Majumdar
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - S S Jha
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - D McGragor
- Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA
| | - S K Banerjee
- 1] Cancer Research Unit, V.A. Medical Center, Kansas City, MO, USA [2] Division of Hematology and Oncology, Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA [3] Department of Anatomy and Cell Biology and Department of Pathology, University of Kansas Medical Center, Kansas City, MO, USA
| |
Collapse
|
7
|
Bravaccini S, Granato AM, Medri L, Foca F, Falcini F, Zoli W, Ricci M, Lanzanova G, Masalu N, Serra L, Buggi F, Folli S, Silvestrini R, Amadori D. Biofunctional characteristics of in situ and invasive breast carcinoma. Cell Oncol (Dordr) 2013; 36:303-10. [PMID: 23807750 DOI: 10.1007/s13402-013-0135-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2013] [Indexed: 02/03/2023] Open
Abstract
PURPOSE The increasing use of breast-conserving surgery makes it essential to identify biofunctional profiles responsible for the progression of in situ to invasive carcinomas to facilitate the detection of lesions that are most likely to relapse or progress and, thus, to be able to offer patients tailored treatment options. Our objective was to analyse and compare biofunctional profiles in ductal carcinomas in situ (DCIS) and invasive ductal carcinomas (IDC). We also aimed to identify markers in tumor and normal surrounding tissues that may be predictive of locoregional recurrence in patients with DCIS. METHODS Biofunctional parameters including mitotic activity, estrogen receptor, progesterone receptor, microvessel density (MVD), c-kit and p27 expression were evaluated in 829 in situ and invasive carcinomas. The impact of the biomarker profiles of DCIS, IDC and normal surrounding tissues on loco-regional recurrence was analyzed. RESULTS A progressive increase in cell proliferation and a concomitant decrease in steroid hormone receptor-positive lesions was observed during the transition from in situ to invasive carcinomas, as also within each subgroup as grade increased. Conversely, p27 expression and MVD dramatically decreased during the transition from in situ to invasive carcinomas. Finally, we found that a low c-kit expression was indicative of IDC relapse. CONCLUSIONS Cell proliferation, hormonal and differentiation characteristics differed in DCIS with respect to IDC, and the main variation in the transition between the two histologic lesions was the decrease in p27 expression and MVD.
Collapse
Affiliation(s)
- Sara Bravaccini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS, Meldola, FC, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
He W, Wang X, Chen L, Guan X. A crosstalk imbalance between p27(Kip1) and its interacting molecules enhances breast carcinogenesis. Cancer Biother Radiopharm 2012; 27:399-402. [PMID: 22690887 DOI: 10.1089/cbr.2010.0802] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
p27(Kip1) (p27) is an inhibitor of cyclin/cyclin-dependent kinase complexes, the nuclear loss of which indicates poor prognoses in various solid tumors. In breast cancer cells, the p27 expression level usually decreases during tumor development and progression. In addition, p27 cytoplasmic mislocalization has been reported, but the exact molecular mechanisms remain unclear. Studies have indicated that its phosphorylation status is the key regulator and that several signal transduction pathways are involved in the regulation of both the expression and distribution of p27. To further understand the signals involved, the differences in the profiles of interacting proteins between tumor and normal cells should be elucidated. It is well known that p27 has various interacting partners, such as cyclin, cyclin-depend kinases, CRM1, Jab1, SKP2, and Spy1. Assays used to profile these proteins show differing intracellular p27 expression and localization depending on the cell-cycle phase. We hypothesize that the imbalance of crosstalk between p27 and the other molecules involved in the same signaling pathways plays an indispensable role in breast cancer carcinogenesis.
Collapse
Affiliation(s)
- Weiwei He
- Department of Oncology, Jinling Hospital, School of Medicine, Nanjing University, China
| | | | | | | |
Collapse
|
9
|
Karst AM, Levanon K, Duraisamy S, Liu JF, Hirsch MS, Hecht JL, Drapkin R. Stathmin 1, a marker of PI3K pathway activation and regulator of microtubule dynamics, is expressed in early pelvic serous carcinomas. Gynecol Oncol 2011; 123:5-12. [PMID: 21683992 PMCID: PMC3171572 DOI: 10.1016/j.ygyno.2011.05.021] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 05/17/2011] [Accepted: 05/18/2011] [Indexed: 11/18/2022]
Abstract
BACKGROUND Most high-grade pelvic serous carcinomas (HGPSCs) arise from fallopian tube epithelium (FTE). To date, few markers have been shown to characterize FTE transformation. Stathmin 1 (STMN1) is a candidate oncogene whose activity is influenced by p53, p27Kip1 (p27), and PI3K/Akt pathway activation. As a microtubule destabilizing protein, STMN1 regulates cytoskeletal dynamics, cell cycle progression, mitosis, and cell migration. This study examines the expression of STMN1 and its negative regulator p27 along the morphologic continuum from normal FTE to invasive carcinoma. METHODS STMN1 and p27 expression were examined by immunohistochemistry (IHC) in benign (n=12) and malignant (n=13) fallopian tubes containing normal epithelium, morphologically benign putative precursor lesions ("p53 signatures"), potential transitional precursor lesions ("proliferative p53 signatures"), tubal intraepithelial carcinoma (TIC), and/or invasive serous carcinoma. STMN1 expression was further assessed in 131 late-stage HGPSCs diagnosed as primary ovarian and in 6 ovarian cancer cell lines by IHC and Western blot, respectively. RESULTS STMN1 expression was absent in benign FTE and infrequently detected in p53 signatures. However, it was weakly expressed in proliferative p53 signatures and robustly induced upon progression to TIC and invasive carcinoma, typically accompanied by decreased p27 levels. STMN1 was expressed in >80% of high-grade serous ovarian carcinomas and cell lines. CONCLUSIONS STMN1 is a novel marker of early serous carcinoma that may play a role in FTE tumor initiation. Our data are consistent with a model by which STMN1 overexpression, resulting from loss of p27-mediated regulation, may potentiate aberrant cell proliferation, migration, and/or loss of polarity during early tumorigenesis.
Collapse
Affiliation(s)
- Alison M. Karst
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Keren Levanon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Sheba Cancer Research Center, Chaim Sheba Medical Center, Ramat Gan, Israel (present affiliation)
| | - Sekhar Duraisamy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Joyce F. Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Michelle S. Hirsch
- Department of Pathology, Division of Women’s and Perinatal Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jonathan L. Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Ronny Drapkin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Pathology, Division of Women’s and Perinatal Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
10
|
Ramljak V, Sučić M, Vrdoljak DV, Borojević N. Expression of Ki-67 and p27(Kip1) in fine-needle aspirates from breast carcinoma and benign breast diseases. Diagn Cytopathol 2011; 39:333-40. [PMID: 21488176 DOI: 10.1002/dc.21391] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cell atypia in breast fine needle aspiration (FNA) can introduce some diagnostic difficulties. Molecules reflecting proliferative cell potential, such as Ki-67 and p27(Kip1) , can help in recognizing the true biological nature of a cell. Thus, the objective of the study was to analyze the difference in Ki-67 and p27(Kip1) cell immunoexpression in breast FNA specimens between fibroadenomas, fibrocystic changes (FCC) with atypia, and breast carcinoma. Microscopic analyses of cell cytomorphology and Ki-67 and p27(Kip1) breast cell immunoexpression were done after standard Pappenheim and immunocytochemical staining (labeled streptavidin-biotin, LSAB) method in autostainer DakoCytomation TechMate™. The study included 50 patients with breast carcinoma, 20 patients with fibroadenoma, and 20 patients with FCC with atypia. High Ki-67 and low or absent p27(Kip1) were found in most patients with breast carcinoma, while majority of FCC with atypia were characterized by low Ki-67 and moderate to high p27(Kip1) cell immunoexpression. Majority of fibroadenomas were associated with low Ki-67 and low to moderate p27(Kip1) cell immunoexpression indicating progressive decrease in cell cycle inhibition, but still not so high proliferative activity as in carcinoma. However, although statistically significant difference for Ki-67 and p27(Kip1) was found between breast lesions in our study, the large ranges observed for each marker make them essentially useless for better cytological diagnosis in a single case. Regarding their opposite role in cell cycle, inverse correlation of Ki-67 and p27(Kip1) was noticed. Poorly differentiated carcinoma cells had mostly high Ki-67 and low p27(Kip1) cell immunoexpression.
Collapse
Affiliation(s)
- Vesna Ramljak
- Department of Cytology, University Hospital for Tumors, Zagreb, Croatia
| | | | | | | |
Collapse
|
11
|
Chu IM, Hengst L, Slingerland JM. The Cdk inhibitor p27 in human cancer: prognostic potential and relevance to anticancer therapy. Nat Rev Cancer 2008; 8:253-67. [PMID: 18354415 DOI: 10.1038/nrc2347] [Citation(s) in RCA: 774] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The cyclin-dependent kinase (Cdk) inhibitor p27 (also known as KIP1) regulates cell proliferation, cell motility and apoptosis. Interestingly, the protein can exert both positive and negative functions on these processes. Diverse post-translational modifications determine the physiological role of p27. Phosphorylation regulates p27 binding to and inhibition of cyclin-Cdk complexes, its localization and its ubiquitin-mediated proteolysis. In cancers, p27 is inactivated through impaired synthesis, accelerated degradation and by mislocalization. Moreover, studies in several tumour types indicate that p27 expression levels have both prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Isabel M Chu
- Braman Family Breast Cancer Institute, and Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, 1580 NW 10th Avenue, Miami, Florida 33136, USA
| | | | | |
Collapse
|
12
|
Millar EKA, Tran K, Marr P, Graham PH. p27KIP-1, cyclin A and cyclin D1 protein expression in ductal carcinoma in situ of the breast: p27KIP-1 correlates with hormone receptor status but not with local recurrence. Pathol Int 2007; 57:183-9. [PMID: 17316413 DOI: 10.1111/j.1440-1827.2007.02079.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Using whole sections of formalin-fixed paraffin-embedded material the expression of p27(KIP-1), cyclin A and cyclin D1 was examined in 60 cases of ductal carcinoma in situ (DCIS) using routine immunohistochemistry with a median follow up of 95 months (range 10-139 months) to identify any association with disease recurrence. Fifty-six patients were treated by local excision and radiotherapy and four by mastectomy without radiotherapy. There was a highly significant positive association between p27(KIP-1) and estrogen receptor/progesterone receptor (ER/PR) status (P = 0.002, P = 0.02) and with p27(KIP-1) and cyclin D1 expression (P = 0.002). A trend between cyclin A and PR status (P = 0.08) was also identified. These findings mirror those described in invasive ductal carcinoma, but there were no associations of any biomarker with histological parameters such as nuclear grade or with local recurrence on univariate analysis, which was present in four of the 56 locally excised group (7.1%). Further examination of a larger cohort may be worthwhile to explore the possible role as adjunctive predictive markers to aid clinical decision making.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Intraductal, Noninfiltrating/metabolism
- Carcinoma, Intraductal, Noninfiltrating/pathology
- Cyclin A/genetics
- Cyclin A/metabolism
- Cyclin D1/genetics
- Cyclin D1/metabolism
- Cyclin-Dependent Kinase Inhibitor p27/genetics
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Middle Aged
- Neoplasm Recurrence, Local
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
Collapse
Affiliation(s)
- Ewan K A Millar
- Department of Anatomical Pathology, South-Eastern Area Laboratory Service, St George Hospital, NSW, Australia.
| | | | | | | |
Collapse
|
13
|
Combined detection of cyclin D1, p27 and DNA content in esophageal cancer. Chin J Cancer Res 2006. [DOI: 10.1007/s11670-006-0282-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
14
|
Dessauvagie BF, Zhao W, Heel-Miller KA, Harvey J, Bentel JM. Characterization of columnar cell lesions of the breast: immunophenotypic analysis of columnar alteration of lobules with prominent apical snouts and secretions. Hum Pathol 2006; 38:284-92. [PMID: 17084437 DOI: 10.1016/j.humpath.2006.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 08/01/2006] [Accepted: 08/02/2006] [Indexed: 11/15/2022]
Abstract
Columnar cell lesions of the breast are detected with increasing frequency in routine pathology practice, in part as a result of the widespread biopsy of nonpalpable breast abnormalities detected by screening mammography. Immunohistochemical investigation of the lesions in relation to the normal breast or to other breast pathologies is not well characterized, and the malignant potential of this spectrum of lesions has not been examined clinically. In this study, a cohort of 45 breast specimens containing columnar cell lesions, in particular, columnar alteration of lobules with prominent apical snouts and secretions (CAPSS), was investigated for expression of a series of breast tumor biomarkers. Using a semiquantitative immunohistochemical scoring system, up-regulation of estrogen, progesterone, and androgen receptors in CAPSS lesions to levels not significantly different from that in in situ or invasive breast tumors was identified. In four cases where CAPSS within a specimen lacked expression of a steroid hormone receptor, the coexisting in situ or invasive carcinoma also lacked expression of that receptor. In 81% of CAPSS lesions, E-cadherin immunostaining was reduced in isolated foci of cells or was decreased in intensity in all cells within the lesion. Quantitation of Ki-67 immunostaining demonstrated that proliferation of cells within CAPSS lesions was increased, compared with normal breast epithelium, but was lower than that detected in in situ or invasive cancers within the same specimens. Results of these analyses indicate that CAPSS shares immunophenotypic alterations with other premalignant lesions, the clinical implications of which may be investigated using established breast tumor biomarkers.
Collapse
Affiliation(s)
- Benjamin F Dessauvagie
- Department of Anatomical Pathology, Royal Perth Hospital, Perth, Western Australia, 6000, Australia
| | | | | | | | | |
Collapse
|
15
|
Mottolese M, Nádasi EA, Botti C, Cianciulli AM, Merola R, Buglioni S, Benevolo M, Giannarelli D, Marandino F, Donnorso RP, Venturo I, Natali PG. Phenotypic changes of p53, HER2, and FAS system in multiple normal tissues surrounding breast cancer. J Cell Physiol 2005; 204:106-12. [PMID: 15622519 DOI: 10.1002/jcp.20275] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
To determine whether phenotypic field changes occur in tissues adjacent to carcinoma, we assayed, by immunohistochemistry, the expression of HER-2, p53, Fas, and FasL in 72 breast cancers (BC) and multiple autologous peritumoral tissues (PTTs) sampled up to 5 cm distance and in 44 benign breast tumors (BBTs). About 5% and 3% of the PTTs and 4.5% and 6.8% of BBTs showed alterations in HER2 and p53 expression, respectively. Of interest, gene amplification was observed in 50% of HER2 positive PTTs, but not in any HER2 positive BBTs. Fas, highly expressed in BBTs and downregulated in BC, maintained its expression in PTTs, whereas FasL, usually negative in BBTs, was upregulated in BC as well as in the PTTs closest (1 cm) to the invasive lesion. Our data suggest that FasL could be a potential novel biomarker of transformation, which may identify, along with HER2 and p53, precursor lesions in a genetically altered breast tissue.
Collapse
|
16
|
Auerkari EI. Methylation of tumor suppressor genes p16(INK4a), p27(Kip1) and E-cadherin in carcinogenesis. Oral Oncol 2005; 42:5-13. [PMID: 15978859 DOI: 10.1016/j.oraloncology.2005.03.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2005] [Accepted: 03/10/2005] [Indexed: 01/14/2023]
Abstract
Not only genomic mutations but also abnormal epigenetic methylation can significantly contribute to gene silencing and carcinogenesis. Methylation is particularly often observed in the CpG islands of the promoter regions in the regulatory genes. However, there are considerable differences in the incidence of methylation e.g. in the tumor suppressor genes, so that aberrant methylation of p16(INK4a) is relatively frequently observed in tumors, p27(Kip1) methylation is rare, and the incidence of E-cadherin methylation occurs at an intermediate rate. Although true genomic defects are generally much less common than methylation, parallel tendencies for both are often observed, probably reflecting the different levels of evolutionary advantage for tumor cells from inactivation of different genes. This also suggests that loss of p27 expression could be more a consequence of carcinogenesis, while lost p16 expression is a true oncogenic event. Due to the role of p27 in maintaining cellular quiescence, however, loss of its expression can still be a useful partial indicator of the aggressiveness of cancer. Loss of E-cadherin or its catenin partners of cellular adhesion will result in increasing invasiveness and metastatic potential of neoplastic cells but, because of several alternative routes to the same effect, incidence of lost expression for one component gene like E-cadherin does not need to be very high. Similarly, there must be a relatively high number of genes with modest or low incidence of aberrant silencing by methylation, to reflect multiple alternatives for the multistep process of carcinogenesis. Nevertheless, methylation of different genes also shows characteristic differences between different cancer and tumor types, and the epigenetic methylation patterns therefore have considerable diagnostic and prognostic potential. Realising this potential requires efficient methods for profiling the status of methylation. Such profiling methods have only recently become available and are still under relatively rapid development.
Collapse
Affiliation(s)
- Elza Ibrahim Auerkari
- Section of Molecular Embryology, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8549, Japan.
| |
Collapse
|
17
|
Ulivi P, Zoli W, Medri L, Amadori D, Saragoni L, Barbanti F, Calistri D, Silvestrini R. c-kit and SCF expression in normal and tumor breast tissue. Breast Cancer Res Treat 2004; 83:33-42. [PMID: 14997053 DOI: 10.1023/b:brea.0000010694.35023.9e] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Several studies have shown a role of the tyrosine kinase receptor, c-kit, and its ligand, SCF, during organogenesis, normal cell development and growth of some tumor histotypes. In breast cancer, studies using different methodologies have shown conflicting results. In the present study we analyzed c-kit and SCF in 14 normal mammary epithelia samples, in 16 in situ and in 75 invasive breast cancers. The expression of c-kit and SCF protein was analyzed by immunohistochemistry and mRNA expression was evaluated by in situ hybridization and reverse-transcriptase polymerase chain reaction (RT-PCR). The different methodologies gave somewhat different results. Using immunohistochemistry and in situ hybridization, protein and mRNA expression of c-kit and SCF were high in normal mammary gland, significantly lower in in situ and almost completely undetectable in invasive breast cancer. Conversely, using RT-PCR, mRNA expression was observed in normal tissue and in all pathologic lesions of mammary gland, probably due to the high sensitivity of the methodology or to the positivity of elements other than tumor cells expressing the receptor and/or its ligand. These results suggest that the c-kit/SCF pathway plays an important role in the maintenance of normal growth of mammary epithelium and that the process of malignant transformation is accompanied by their progressive loss. Furthermore, we demonstrated that different results are attributable to different methodologies and that morphologic approaches are the most reliable for defining the cellular source of c-kit or SCF expression.
Collapse
Affiliation(s)
- Paola Ulivi
- Department of Medical Oncology, Forlì, Italy
| | | | | | | | | | | | | | | |
Collapse
|
18
|
LaRue KEA, Khalil M, Freyer JP. Microenvironmental regulation of proliferation in multicellular spheroids is mediated through differential expression of cyclin-dependent kinase inhibitors. Cancer Res 2004; 64:1621-31. [PMID: 14996720 DOI: 10.1158/0008-5472.can-2902-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Multicellular spheroids composed of transformed cells are known to mimic the growth characteristics of tumors and to develop gradients in proliferation with increasing size. This progressive accumulation of quiescent cells is presumably an active process that occurs in response to the microenvironmental stresses that develop within the three-dimensional structure, and, yet, little is known regarding either the signals that induce the cell cycle arrest or the molecular basis for the halt in proliferation. We have previously reported that regulation of cyclin-dependent kinase (CDK) inhibitors (CKIs) differs in monolayer versus spheroid cell culture. In this study, we have examined the expression of three CKIs in EMT6 mouse mammary carcinoma and MEL28 human melanoma spheroids, as a function both of spheroid size and of location within the spheroid. We report that expression of the CKIs p18(INK4c), p21(waf1/cip1), and p27(Kip1) all increase as the spheroid grows and develops a quiescent cell fraction. However, by examining protein expression in discrete regions of the spheroid, we have found that only p18(INK4c) and p27(Kip1) expression positively correlate with growth arrest, whereas p21(waf1/cip1) is expressed predominantly in proliferating cells. Further analysis indicated that, in the quiescent cells, p18(INK4c) is found in increasing association with CDK6, whereas p27(Kip1) associates predominantly with CDK2. In MEL28 cells, CDK2 activity is completely abrogated in the inner regions of the spheroid, whereas in EMT6 cells, CDK2 activity decreases in accordance with a decrease in expression. We also observed a decrease in all cell cycle regulatory proteins in the innermost spheroid fraction, including CDKs, CKIs, and cyclins. Induction of CKIs from separate families, as well as their association with distinct target CDKs, suggests that there may be multiple checkpoints activated to ensure cell cycle arrest in non-growth-conducive environments. Furthermore, because very similar observations were made in both a human melanoma cell line and a mouse mammary carcinoma cell line, our results indicate that these checkpoints, as well as the signal transduction pathways that activate them, are highly conserved.
Collapse
Affiliation(s)
- Karen E A LaRue
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | | | | |
Collapse
|
19
|
Alkarain A, Jordan R, Slingerland J. p27 deregulation in breast cancer: prognostic significance and implications for therapy. J Mammary Gland Biol Neoplasia 2004; 9:67-80. [PMID: 15082919 DOI: 10.1023/b:jomg.0000023589.00994.5e] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
p27 is a key regulator of G1-to-S phase progression. It prevents premature activation of cyclin E-cdk2 in G1 and promotes the assembly and activation of D-type cyclin-cdks. While the p27 gene is rarely mutated in human cancers, the action of p27 is impaired in breast and other human cancers through accelerated p27 proteolysis, sequestration by cyclin D-cdks, and by p27 mislocalization in tumor cell cytoplasm. Reduced p27 protein is strongly associated with high histopathologic tumor grade, reflecting a lack of tumor differentiation. Loss of p27 is also an indicator of poor patient outcome in a majority of breast cancer studies, including node negative disease. The broad application of p27 in the clinical evaluation of breast cancer prognosis will require a consensus on methods of tumor fixation, staining, and scoring. This review will focus on mechanisms of p27 regulation in normal cells and how deregulation of p27 may arise in breast and other human cancers. The prognostic significance of p27 in human breast cancer and the possible therapeutic implications of these findings will also be reviewed.
Collapse
Affiliation(s)
- A Alkarain
- Molecular and Cell Biology, Sunnybrook and Women's Health Sciences Centre, University of Toronto, Bayview Avenue, Toronto, Ontario, Canada
| | | | | |
Collapse
|
20
|
Troncone G, Migliaccio I, Caleo A, Palmieri EA, Iaccarino A, Sparano L, Vetrani A, Palombini L. p27Kip1 Expression and grading of breast cancer diagnosed on cytological samples. Diagn Cytopathol 2004; 30:375-80. [PMID: 15176022 DOI: 10.1002/dc.20059] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The progressive reduction in p27(Kip1) (p27) protein immunohistochemical staining with increasing histological grading is a well-established finding occurring in breast cancer, and its role as diagnostic complement and prognostic marker has been thoroughly evaluated. To clarify whether this test may be applied to breast cytopathology, we performed p27 immunostaining on fresh fine-needle cytology (FNC) samples from 10 benign and 40 malignant breast lesions. On average, p27 immunostaining was significantly lower in carcinomas than in benign lesions (P < 0.005). In particular, among carcinomas, p27 immunostaining progressively reduced from well-to poorly differentiated lesions (G1 vs. G2, P < 0.05; G1 vs. G3, P < 0.001; G2 vs. G3; P < 0.001). A similar trend was noted in a subgroup of 20 matched FNCs and histological samples of breast carcinomas, when p27 immunostaining on FNCs was stratified according to the histological grading (G1 vs. G2, P = 0.18; G1 vs. G3, P < 0.05; G2 vs. G3, P < 0.05). In addition, p27 immunostaining on FNCs showed a good positive correlation with that on histology (Spearman R = 0.58; P < 0.01), with a diagnostic concordance between samples of 85%, by using the standard 50% positive cell cutoff. Taken in concert, our data suggest that p27 immunostaining is a reliable marker of tumor cell differentiation in breast cytopathology as well as in histopathology. Accordingly, staining FNCs for p27 may be an useful complement in addition to cytological grading in the preoperative assessment of breast cancer.
Collapse
Affiliation(s)
- Giancarlo Troncone
- Dipartimento di Scienze Biomorfologiche e Funzionali, University Federico II School of Medicine, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Davison EA, Lee CSL, Naylor MJ, Oakes SR, Sutherland RL, Hennighausen L, Ormandy CJ, Musgrove EA. The cyclin-dependent kinase inhibitor p27 (Kip1) regulates both DNA synthesis and apoptosis in mammary epithelium but is not required for its functional development during pregnancy. Mol Endocrinol 2003; 17:2436-47. [PMID: 12933906 DOI: 10.1210/me.2003-0199] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Decreased expression of the cyclin-dependent kinase (CDK) inhibitor p27(Kip1) is common in breast cancer and is associated with poor prognosis. p27 is also an important mediator of steroidal regulation of cell cycle progression. We have therefore investigated the role of p27 in mammary epithelial cell proliferation. Examination of the two major functions of p27, assembly of cyclin D1-Cdk4 complexes and inhibition of Cdk2 activity, revealed that cyclin D1-Cdk4 complex formation was not impaired in p27-/- mammary epithelial cells in primary culture. However, cyclin E-Cdk2 activity was increased approximately 3-fold, indicating that the CDK inhibitory function of p27 is important in mammary epithelial cells. Increased epithelial DNA synthesis was observed during pregnancy in p27-/- mammary gland transplants, but this was paralleled by increased apoptosis. During pregnancy and at parturition, development and differentiation of p27+/+ and p27-/- mammary tissue were indistinguishable. These results demonstrate a role for p27 in both the proliferation and survival of mammary epithelial cells. However, the absence of morphological and cellular defects in p27-/- mammary tissue during pregnancy raises the possibility that loss of p27 in breast cancer may not confer an overall growth advantage unless apoptosis is also impaired.
Collapse
Affiliation(s)
- Elizabeth A Davison
- Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, New South Wales 2010, Australia
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Alkarain A, Slingerland J. Deregulation of p27 by oncogenic signaling and its prognostic significance in breast cancer. Breast Cancer Res 2003; 6:13-21. [PMID: 14680481 PMCID: PMC314445 DOI: 10.1186/bcr722] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
p27 is a key regulator of progression from G1 to S phase. Although the gene encoding p27 is rarely mutated in human cancers, p27 is functionally inactivated in a majority of human cancers through accelerated p27 proteolysis, through sequestration by cyclin D-cyclin-dependent kinase complexes and by cytoplasmic mislocalization. Here we review mechanisms whereby oncogenic activation of receptor tyrosine kinase and Ras pathways lead to accelerated p27 proteolysis and p27 mislocalization in cancer cells. The prognostic significance of p27 in human breast cancer is also reviewed.
Collapse
Affiliation(s)
- Angel Alkarain
- Sunnybrook and Women's Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Joyce Slingerland
- Braman Breast Cancer Institute, University of Miami School of Medicine, Miami, FL, USA
| |
Collapse
|