scholarly journals Differential values of ki-67 index and mitotic index of proliferating cell population. An assessment of cell cycle and prognosis in radiation therapy for cervical cancer

Cancer ◽  
1993 ◽  
Vol 72 (8) ◽  
pp. 2401-2408 ◽  
Author(s):  
Takashi Nakano ◽  
Kuniyuki Oka
1971 ◽  
Vol 48 (2) ◽  
pp. 248-252 ◽  
Author(s):  
S. H. Socher ◽  
D. Davidson

Treatment of Vicia faba lateral roots with a range of concentrations of 5-aminouracil (5-AU) indicate that cells are stopped at a particular point in interphase. The timing of the fall in mitotic index suggests that cells are held at the S - G2 transition. When cells are held at this point, treatments with 5-AU can be used to estimate the duration of G2 + mitosis/2 of proliferating cells. Treatment with 5-AU can also be used to demonstrate the presence of subpopulations of dividing cells that differ in their G2 duration. Using this method, 5-AU-induced inhibition, we have confirmed that in V. faba lateral roots there are two populations of dividing cells: (a) a fast-dividing population, which makes up ∼85% of the proliferating cell population and has a G2 + mitosis/2 duration of 3.3 hr, and (b) a slow-dividing population, which makes up ∼15% of dividing cells and has a G2 duration in excess of 12 hr. These estimates are similar to those obtained from percentage labeled mitosis (PLM) curves after incorporation of thymidine-3H.


2020 ◽  
Vol 20 (1) ◽  
Author(s):  
Chen Wang ◽  
Shiqing Shao ◽  
Li Deng ◽  
Shelian Wang ◽  
Yongyan Zhang

Abstract Background Radiation resistance is a major obstacle to the prognosis of cervical cancer (CC) patients. Many studies have confirmed that long non-coding RNAs (lncRNAs) are involved in the regulation of radiosensitivity of cancers. However, whether small nucleolar RNA host gene 12 (SNHG12) regulates the radiosensitivity of CC remains unknown. Methods Quantitative real-time polymerase chain reaction was used to measure the expression levels of SNHG12 and microRNA-148a (miR-148a). The radiosensitivity of cells was evaluated by clonogenic assay. Flow cytometry and caspase-3 activity assay were performed to assess the apoptosis ability and cell cycle distribution of cells. Besides, dual-luciferase reporter and RNA immunoprecipitation assay were used to verify the interaction between miR-148a and SNHG12 or cyclin-dependent kinase 1 (CDK1). Also, the protein levels of CDK1, CCND1 and γ-H2AX were detected by western blot analysis. Furthermore, in vivo experiments were conducted to verify the effect of SNHG12 on CC tumor growth. Ki-67 and TUNEL staining were employed to evaluate the proliferation and apoptosis rates in vivo. The hematoxylin and eosin (HE) staining were employed to evaluate the tumor cell morphology. Results SNHG12 was upregulated in CC tissues and cells, and its knockdown improved the radiosensitivity by promoting the radiation-induced apoptosis and cell cycle arrest of CC cells. Also, miR-148a could be sponged by SNHG12 and could target CDK1. MiR-148a inhibitor or CDK1 overexpression could invert the promotion effect of silenced-SNHG12 on CC radiosensitivity. Meanwhile, SNHG12 interference reduced the tumor growth of CC, increased miR-148a expression, and inhibited CDK1 level in vivo. Conclusion LncRNA SNHG12 promoted CDK1 expression to regulate the sensitivity of CC cells to radiation through sponging miR-148a, indicating that SNHG12 could be used as a potential biomarker to treat the radiotherapy resistance of CC patients.


2000 ◽  
Vol 124 (10) ◽  
pp. 1457-1462
Author(s):  
Keith F. Izban ◽  
Serhan Alkan ◽  
Timothy P. Singleton ◽  
Eric D. Hsi

Abstract Background.—Mantle cell lymphoma (MCL) is characterized by overexpression of cyclin D1, a G1 cyclin that participates in the control of cell cycle progression at the G1 to S phase transition. In addition to cyclin D1, other cell cycle regulatory molecules may be involved in the proliferation and progression of MCL. Mutation of p53, deletion of p16INK4a, and loss of p21WAF1 expression have been reported in some cases of blastoid MCL. Objective.—We sought to examine levels of expression of these proteins in typical and blastoid MCL and to determine whether differences were present between these subtypes of lymphomas. Design.—A retrospective series of typical and blastoid MCLs was evaluated for expression of the cell cycle–related proteins cyclin D1, p21WAF1, p27KIP1, Ki-67, and p53, as well as mitotic index. Paraffin-embedded archival tissues from 24 MCL specimens (17 typical, 7 blastoid) were immunostained with antibodies to p21WAF1, p27KIP1, p53, Ki-67, and cyclin D1. The percentage of positive cells for each specimen was estimated by counting 1500 cells under oil immersion microscopy. Levels of antigen expression were compared for the typical and blastoid MCLs. The mitotic index was estimated using twenty 100× oil immersion fields (OIFs) for each specimen. Results.—Cyclin D1 expression was seen in 22/24 specimens (92%). Blastoid MCLs were characterized by a significantly higher mean mitotic index (>20 mitoses/20 OIFs) and Ki-67 index (>45%) when compared with typical MCLs (P < .001 and P < .008, respectively; Fisher's exact test). High expression of p27KIP1 (>25% staining) was seen more frequently in typical MCLs than in the blastoid variants (P = .03; Fisher's exact test). No significant differences were found between typical and blastoid MCLs for the expression of p21WAF1 or p53. Conclusions.—A significantly higher mitotic index and Ki-67 index were found in blastoid MCLs as compared with typical MCLs. Low p27KIP1 expression was associated with the blastoid MCL variant. These findings confirm the high proliferative nature of blastoid MCL and suggest a role for p27KIP1 in the negative regulation of the cell cycle in MCL.


Cancer ◽  
2009 ◽  
Vol 115 (9) ◽  
pp. 1875-1882 ◽  
Author(s):  
Yoshiyuki Suzuki ◽  
Kuniyuki Oka ◽  
Tatsuya Ohno ◽  
Shingo Kato ◽  
Hirohiko Tsujii ◽  
...  

2012 ◽  
Vol 30 (4_suppl) ◽  
pp. 242-242
Author(s):  
Ben Lawrence ◽  
Simon Schimmack ◽  
Bernhard Svejda ◽  
Ignat Drozdov ◽  
Daniele Alaimo ◽  
...  

242 Background: Ki-67 is the major proliferative marker in clinical use to determine neuroendocrine neoplasm (NEN) prognosis. Ki-67 is unable to predict the outcome of SI-NENs, as the majority have a low (≤2) Ki-67%. Therefore, we aimed to identify a sensitive panel of proliferative markers using qRT-PCR to more accurately define the proliferation of these slow growing tumors. Methods: We identified genes with a mechanistic function in cell cycle progression that were over-expressed in RNA microarrays of SI-NENs (n=8) compared to adjacent normal tissue (n=4) (dCHIP, annotation databases). Timing of marker gene expression (qRT-PCR) in proliferating cell-cycle phases (S, G2, M) was determined in flow-sorted SI-NEN cell lines (KRJ-1, H-STS) after propidium iodide staining. RNA expression of candidate proliferative markers was then investigated using an in vivo model and two independent tumor datasets, and transcript level compared to Ki-67% protein expression (immunohistochemical staining). Results: Twenty genes with a mechanistic role in proliferation were identified and 17 confirmed to be expressed in proliferating cell cycle phases. Each tumor expressed a unique profile of the 17 proliferative markers. Both Ki-67 protein and Ki-67 RNA transcript levels failed to differentiate in vivo SI-NEN models or patient samples despite variable proliferative capacity (e.g., WDNETs versus WDNECs). Although most tumors showed low levels of Ki-67 expression, the tumors expressed high levels of select alternative proliferative markers. Hierarchical clustering provided a novel and clinically meaningful prognostic classification. Conclusions: Proliferation of individual SI-NENs is regulated by unique combinations of multiple genes with a mechanistic role in cell-cycle progression. Regulation of proliferation in SI-NENs is therefore complex and cannot accurately be defined by Ki-67 as a single marker. A panel of proliferative RNA markers has potential to significantly improve prognostication in patients with SI-NENs.


Endocrinology ◽  
2012 ◽  
Vol 153 (10) ◽  
pp. 5011-5022 ◽  
Author(s):  
Yoel Toledano ◽  
Svetlana Zonis ◽  
Song-Guang Ren ◽  
Kolja Wawrowsky ◽  
Vera Chesnokova ◽  
...  

Abstract Because pregnancy and estrogens both induce pituitary lactotroph hyperplasia, we assessed the expression of pituitary cell cycle regulators in two models of murine pituitary hyperplasia. Female mice were assessed during nonpregnancy, pregnancy, day of delivery, and postpartum. We also implanted estradiol (E2) pellets in female mice and studied them for 2.5 months. Pituitary weight in female mice increased 2-fold after E2 administration and 1.4-fold at day of delivery, compared with placebo-treated or nonpregnant females. Pituitary proliferation, as assessed by proliferating cell nuclear antigen and/or Ki-67 staining, increased dramatically during both mid-late pregnancy and E2 administration, and lactotroph hyperplasia was also observed. Pregnancy induced pituitary cell cycle proliferative and inhibitory responses at the G1/S checkpoint. Differential cell cycle regulator expression included cyclin-dependent kinase inhibitors, p21Cip1, p27Kip1, and cyclin D1. Pituitary cell cycle responses to E2 administration partially recapitulated those effects observed at mid-late pregnancy, coincident with elevated circulating mouse E2, including increased expression of proliferating cell nuclear antigen, Ki-67, p15INK4b, and p21Cip1. Nuclear localization of pituitary p21Cip1 was demonstrated at mid-late pregnancy but not during E2 administration, suggesting a cell cycle inhibitory role for p21Cip1 in pregnancy, yet a possible proproliferative role during E2 administration. Most observed cell cycle protein alterations were reversed postpartum. Murine pituitary meets the demand for prolactin during lactation associated with induction of both cell proliferative and inhibitory pathways, mediated, at least partially, by estradiol.


2017 ◽  
Vol 27 (4) ◽  
pp. 628-633 ◽  
Author(s):  
Yuzhao Zhang ◽  
Hongyi Gao ◽  
Xiang Gao ◽  
Senlin Huang ◽  
Kunhe Wu ◽  
...  

BackgroundCervical cancer is one of the most common cancers in women worldwide. Emerging evidence suggests that kin17 is a tumor-promoting protein in some types of solid tumors. However, whether kin17 contributes to cervical cancer carcinogenesis remains unknown.MethodsKin17 expression in clinical samples from Guangdong Women and Children's Hospital and Health Institute was detected by immunohistochemical staining. A series of functional experiments including 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay, 5-bromo-2′-deoxyuridine assay, colony formation, transwell assay, flow cytometry of apoptosis, and cell cycle were performed to explore the roles of kin17 in cervical cancer cells HeLa.ResultsIn this study, we showed for the first time that the expression of kin17 was significantly increased in clinical cervical cancer samples, and associated with tumor differentiation, lymph node metastasis, and ki-67 expression in a clinicopathologic characteristics review. Furthermore, silence of kin17 in HeLa cells inhibited cell proliferation, clone formation, cell cycle progression, migration, and invasion, and also promoted cell apoptosis.ConclusionOur findings demonstrate that kin17 is closely related to the cell proliferation and invasion of cervical cancer and could be a novel diagnostic and therapeutic target for cervical cancer management. The underlying mechanisms should be elucidated in future research.


Sign in / Sign up

Export Citation Format

Share Document