1.Driving effect of P16 methylation on telomerase reverse transcriptase-mediated immortalization and transformation of normal human fibroblasts.
Xuehong ZHANG ; Paiyun LI ; Ying GAN ; Shengyan XIANG ; Liankun GU ; Jing ZHOU ; Xiaorui ZHOU ; Peihuang WU ; Baozhen ZHANG ; Dajun DENG
Chinese Medical Journal 2025;138(3):332-342
BACKGROUND:
P16 inactivation is frequently accompanied by telomerase reverse transcriptase ( TERT ) amplification in human cancer genomes. P16 inactivation by DNA methylation often occurs automatically during immortalization of normal cells by TERT . However, direct evidence remains to be obtained to support the causal effect of epigenetic changes, such as P16 methylation, on cancer development. This study aimed to provide experimental evidence that P16 methylation directly drives cancer development.
METHODS:
A zinc finger protein-based P16 -specific DNA methyltransferase (P16-Dnmt) vector containing a "Tet-On" switch was used to induce extensive methylation of P16 CpG islands in normal human fibroblast CCD-18Co cells. Battery assays were used to evaluate cell immortalization and transformation throughout their lifespan. Cell subcloning and DNA barcoding were used to track the diversity of cell evolution.
RESULTS:
Leaking P16-Dnmt expression (without doxycycline-induction) could specifically inactivate P16 expression by DNA methylation. P16 methylation only promoted proliferation and prolonged lifespan but did not induce immortalization of CCD-18Co cells. Notably, cell immortalization, loss of contact inhibition, and anchorage-independent growth were always prevalent in P16-Dnmt&TERT cells, indicating cell transformation. In contrast, almost all TERT cells died in the replicative crisis. Only a few TERT cells recovered from the crisis, in which spontaneous P16 inactivation by DNA methylation occurred. Furthermore, the subclone formation capacity of P16-Dnmt&TERT cells was two-fold that of TERT cells. DNA barcoding analysis showed that the diversity of the P16-Dnmt&TERT cell population was much greater than that of the TERT cell population.
CONCLUSION
P16 methylation drives TERT -mediated immortalization and transformation of normal human cells that may contribute to cancer development.
Humans
;
Telomerase/genetics*
;
DNA Methylation/physiology*
;
Fibroblasts/cytology*
;
Cyclin-Dependent Kinase Inhibitor p16/metabolism*
;
Cell Line
;
Cell Transformation, Neoplastic/genetics*
2.Cyclin D1 immunohistochemical stain as adjunct immunomarker in CD99 positive malignant small round blue cell tumors with primary consideration of Primitive Neuroectodermal Tumor/Ewing Sarcoma in a pediatric tertiary hospital
Marie Janina Gail B. Co ; Ivy Marie Viola-Cruz
The Philippine Children’s Medical Center Journal 2025;21(1):18-26
OBJECTIVES:
This study aimed to evaluate the diagnostic accuracy of Cyclin D1 as an adjunct immunomarker in CD99 positive small round cell neoplasms with primary consideration of PNET/EWS.
MATERIALS AND METHODS:
Tissue from 2017 to 2023 with a histopathologic diagnosis of CD99 positive small round blue cell tumors with primary consideration of Primitive Neuroectodermal Tumor (PNET)/Ewing Sarcoma were retrieved and Cyclin D1 immunohistochemical staining done. Diagnostic accuracy of Cyclin D1 immunostaining was determined by calculating the sensitivity, specificity, positive predictive value, and negative predictive value.
RESULTS:
Cyclin D1 immunohistochemical staining was performed in 19 specimens available, of which 13 yielded a positive result. Of these, 8 had a final histopathologic diagnosis of CD99 positive small round blue cell tumor with primary consideration of PNET/Ewing Sarcoma, resulting in sensitivity of 61.54%, specificity of 100%, positive predictive value of 100% and negative predictive value of 50.0%. The overall accuracy is 72.2%.
CONCLUSION
Cyclin D1 can be used as an adjunct immunomarker to aid in the diagnosis of CD99 positive round cell tumor with primary consideration of PNET/Ewing Sarcoma specifically in resource limited settings where molecular testing is not readily available. Given the high specificity of Cyclin D1 in such cases, it can be used to rule out other small round blue cell tumors that can also stain positive for CD99 such as Rhabdomyosarcoma. However, interpretation must be done in conjunction with the results of other immunohistochemical stains in order to increase its diagnostic accuracy.
Human
;
Male,Female
;
Cells
;
Sarcoma, Ewing
;
Sarcoma
;
Neuroectodermal tumors, Primitive
;
Cyclin D1
3.Targeting WEE1: a rising therapeutic strategy for hematologic malignancies.
Hao-Bo LI ; Thekra KHUSHAFA ; Chao-Ying YANG ; Li-Ming ZHU ; Xing SUN ; Ling NIE ; Jing LIU
Acta Physiologica Sinica 2025;77(5):839-854
Hematologic malignancies, including leukemia, lymphoma, and multiple myeloma, are hazardous diseases characterized by the uncontrolled proliferation of cancer cells. Dysregulated cell cycle resulting from genetic and epigenetic abnormalities constitutes one of the central events. Importantly, cyclin-dependent kinases (CDKs), complexed with their functional partner cyclins, play dominating roles in cell cycle control. Yet, efforts in translating CDK inhibitors into clinical benefits have demonstrated disappointing outcomes. Recently, mounting evidence highlights the emerging significance of WEE1 G2 checkpoint kinase (WEE1) to modulate CDK activity, and correspondingly, a variety of therapeutic inhibitors have been developed to achieve clinical benefits. Thus, WEE1 may become a promising target to modulate the abnormal cell cycle. However, its function in hematologic diseases remains poorly elucidated. In this review, focusing on hematologic malignancies, we describe the biological structure of WEE1, emphasize the latest reported function of WEE1 in the carcinogenesis, progression, as well as prognosis, and finally summarize the therapeutic strategies by targeting WEE1.
Humans
;
Protein-Tyrosine Kinases/physiology*
;
Hematologic Neoplasms/drug therapy*
;
Cell Cycle Proteins/antagonists & inhibitors*
;
Nuclear Proteins/antagonists & inhibitors*
;
Cyclin-Dependent Kinases
;
Molecular Targeted Therapy
;
Animals
4.Research progress on copy number alterations in pediatric B-cell acute lymphoblastic leukemia.
Chinese Journal of Contemporary Pediatrics 2025;27(6):746-752
Copy number alteration (CNA) is a significant genetic change in pediatric B-cell acute lymphoblastic leukemia (B-ALL), with <i>CDKN2A/Bi> deletions, <i>PAX5i> deletions, and <i>IKZF1i> deletions being the most common. Recent studies have increasingly highlighted the potential prognostic significance of these gene deletions and multiple co-deletions in pediatric B-ALL. This paper reviews the main detection methods for CNA, as well as the prognostic characteristics and treatment approaches for common CNA in pediatric B-ALL.
Humans
;
DNA Copy Number Variations
;
Child
;
PAX5 Transcription Factor/genetics*
;
Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics*
;
Cyclin-Dependent Kinase Inhibitor p15/genetics*
;
Ikaros Transcription Factor/genetics*
;
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics*
;
Gene Deletion
;
Cyclin-Dependent Kinase Inhibitor p16/genetics*
;
Prognosis
5.Beckwith-Wiedemann syndrome with ganglioneuroblastoma: a case report.
Jin-Fa TOU ; Ci-Yuan FENG ; Bin XU ; Jing-Jing YE
Chinese Journal of Contemporary Pediatrics 2025;27(8):1022-1026
This paper reports the case of a 10-month-old male infant with Beckwith-Wiedemann syndrome (BWS) who presented with a reducible right inguinal mass and an empty scrotum for 10 months and was admitted for elective surgery. Preoperative ultrasonography revealed a right adrenal mass, which was pathologically diagnosed as ganglioneuroblastoma (GNB) after surgical excision. The patient exhibited characteristic features of BWS, including omphalocele, flame-shaped nevus on the forehead, bilateral earlobe creases, and embryonal tumor. Next-generation sequencing identified a heterozygous mutation in the <i>CDKN1Ci> gene (chr11:2905365), confirming the diagnosis of BWS. Early diagnosis, standardized management, and tumor surveillance are crucial for improving prognosis in children with BWS. Ultrasonography enables early detection of tumors and informs clinical decision-making regarding intervention.
Humans
;
Beckwith-Wiedemann Syndrome/genetics*
;
Male
;
Ganglioneuroblastoma/complications*
;
Infant
;
Cyclin-Dependent Kinase Inhibitor p57/genetics*
;
Mutation
6.Prognostic Value of <i>CDKN2Ai> Copy Number Deletion in Patients with Diffuse Large B-Cell Lymphoma.
Wei-Yuan MA ; Le-Tian SHAO ; Wen-Xin TIAN ; Sha LIU ; Yan LI
Journal of Experimental Hematology 2025;33(2):379-386
OBJECTIVE:
To investigate the relationship between <i>CDKN2Ai> copy number deletion and clinical features of patients with diffuse large B-cell lymphoma (DLBCL) and its prognostic value.
METHODS:
155 newly diagnosed DLBCL patients with complete clinical data in the Department of Hematology of People's Hospital of Xinjiang Uygur Autonomous Region from March 2009 to March 2022 were included, formalin-fixed paraffin-embedded tumor tissues were obtained and DNA was extracted from them, and next-generation sequencing technology was applied to target sequencing including 475 lymphoma-related genes, the relationship between <i>CDKN2Ai> copy number deletion and clinical features, high-frequency mutated genes and overall survival (OS) of DLBCL patients were analyzed.
RESULTS:
<i>CDKN2Ai> copy number deletion was present in 12.9% (20/155) of 155 DLBCL patients, grouped according to the presence or absence of copy number deletion of <i>CDKN2Ai>, and a higher proportion of patients with IPI≥3 were found in the <i>CDKN2Ai> copy number deletion group compared to the group with no <i>CDKN2Ai> copy number deletion (80% <i>vsi> 51.5%, <i>Pi> =0.015) and were more likely to have bulky disease (20% <i>vsi> 5.2%, <i>Pi> =0.037). Survival analysis showed that the 5-year OS of patients in the <i>CDKN2Ai> copy number deletion group was significantly lower than that of the non-deletion group (51.3% <i>vsi> 69.2%, <i>Pi> =0.047). Multivariate Cox analysis showed that IPI score≥3 (<i>Pi> =0.007), <i>TP53i> mutation (<i>Pi> =0.009), and <i>CDKN2Ai> copy number deletion (<i>Pi> =0.04) were independent risk factors affecting the OS of DLBCL patients.
CONCLUSION
<i>CDKN2Ai> copy number deletion is an independent risk factor for OS in DLBCL, and accurate identification of <i>CDKN2Ai> copy number deletion can predict the prognosis of DLBCL patients.
Humans
;
Lymphoma, Large B-Cell, Diffuse/genetics*
;
Prognosis
;
Cyclin-Dependent Kinase Inhibitor p16/genetics*
;
DNA Copy Number Variations
;
Female
;
Male
;
Middle Aged
;
Gene Deletion
;
Adult
;
Aged
7.Impacts of Sulforaphane on Cell Proliferation and Apoptosis in Acute Promyelogenous Leukemia by Regulating the PI3K/Akt/mTOR Signaling Pathway.
Cui-Cui WANG ; Zhen-Jing LI ; Xiu-Hong JIA ; Jian-Chang LI
Journal of Experimental Hematology 2025;33(3):633-639
OBJECTIVE:
To investigate the impacts of sulforaphane (SPN) on cell proliferation and apoptosis in acute promyelogenous leukemia by regulating the PI3K/Akt/mTOR signaling pathway.
METHODS:
NB4 cells were divided into 5 μmol/L SPN group, 10 μmol/L SPN group, 20 μmol/L SPN group, 740 Y-P (10 μmol/L) group and 20 μmol/L SPN+740 Y-P group, and the untreated NB4 cells were used as the control group. CCK-8, Hoechst 33342 staining, flow cytometry and monodansulfonylpentanediamine (MDC) were used to detect cell proliferation, apoptosis and autophagy, respectively. The expression levels of <i>Bcl-2, Bax, cyclin D1i> and <i>LC3Bi> mRNA were detected by qRT-PCR. Western blot was used to detect the expression levels of PI3K/Akt/mTOR pathway-related proteins in NB4 cells.
RESULTS:
Compared with the control group, the proliferation rate, <i>Bcl-2, cyclin D1i> mRNA expressions, p-PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR ratio were greatly increased in the 740 Y-P group (<i>Pi> < 0.05), the apoptosis rate, percentage of MDC positive, <i>Baxi> and <i>LC3Bi> mRNA expression levels were greatly decreased (<i>Pi> < 0.05). The proliferation rate, <i>Bcl-2, cyclin D1i> mRNA expression levels, p-PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR ratio were greatly decreased in the 5 μmol/L SPN group, 10 μmol/L SPN group, and 20 μmol/L SPN group (<i>Pi> < 0.05), the apoptosis rate, percentage of MDC positive,<i>Baxi> and <i>LC3Bi> mRNA expression levels were greatly increased, there were differences among different SPN treatment groups (<i>Pi> < 0.05). Compared with the 20 μmol/L SPN group, the proliferation rate, <i>Bcl-2, cyclin D1i> mRNA expression levels, p-PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR ratio were greatly increased in the 20 μmol/L SPN+740 Y-P group(<i>Pi> < 0.05), the apoptosis rate, percentage of MDC positive, <i>Baxi> and <i>LC3Bi> mRNA expression levels were greatly decreased (<i>Pi> < 0.05). Compared with the 740 Y-P group, the proliferation rate, <i>Bcl-2, cyclin D1i> mRNA expression levels, p-PI3K/PI3K, p-Akt/Akt, and p-mTOR/mTOR ratio in the 20 μmol/L SPN+740 Y-P group were greatly reduced (<i>Pi> < 0.05), the apoptosis rate, percentage of MDC positive, <i>Baxi> and <i>LC3Bi> mRNA expression levels were greatly increased (<i>Pi> < 0.05).
CONCLUSION
SPN reduces the proliferation of acute promyelocytic leukemia cells and promotes cells apoptosis by inhibiting the PI3K/Akt/mTOR signaling pathway.
Cell Proliferation/drug effects*
;
Apoptosis/drug effects*
;
Humans
;
TOR Serine-Threonine Kinases/metabolism*
;
Signal Transduction/drug effects*
;
Proto-Oncogene Proteins c-akt/metabolism*
;
Isothiocyanates/pharmacology*
;
Phosphatidylinositol 3-Kinases/metabolism*
;
Sulfoxides
;
Cell Line, Tumor
;
Cyclin D1/metabolism*
8.Hydroxysafflor Yellow A Ameliorates the Replicative Senescence of Human Umbilical Cord Mesenchymal Stem Cells by Suppressing Oxidative Stress.
Si-Yun WANG ; Qi ZHU ; Chun-Xia TAN ; Fang LU ; Tao LU
Journal of Experimental Hematology 2025;33(5):1507-1515
OBJECTIVE:
To investigate the effects and mechanisms of hydroxysafflor yellow A (HSYA) on replicative senescence in human umbilical cord mesenchymal stem cells (hUC-MSCs).
METHODS:
hUC-MSCs were cultured to construct a replicative senescence model through continuous amplification <i>in vitroi>. Cells at passage 2 served as the control group, while cells at passage 10 were designated as the senescence group. The senescent cells were cultured in a culture medium containing HSYA. Cell viability was detected by the CCK-8 assay, and cell confluence was analyzed using the Incucyte S3 live-cell analysis system. The optimal concentration and time point were determined and utilized for subsequent experiments. Senescent cells were pretreated with 0.01 mg/ml HSYA, and the proportion of senescence-associated β-galactosidase (SA-β-gal) positive cells was detected to assess the senescence state. The relative telomere length was detected by qPCR. Reactive oxygen species (ROS) levels were measured using the fluorescent probe DCFH-DA. Mitochondrial membrane potential was assessed by JC-1 staining. The expression of <i>p53, p16, p21, OCT4i>, and <i>SOX2i> genes was detected by qPCR. The expression of p16, p53, OCT4, and SOX2 proteins was analyzed by Western blot.
RESULTS:
HSYA significantly decreased the SA-β-gal positive staining rate, inhibited telomere attrition, reduced the ROS accumulation, increased mitochondrial membrane potential in senescent cells. Additionally, HSYA downregulated the expression of <i>p53i> and <i>p16i>, and upregulated the expression of <i>OCT4i>. HSYA decreased p16 protein level and increased OCT4 and SOX2 protein levels.
CONCLUSION
HSYA may ameliorate replicative senescence in hUC-MSCs by modulating the p53 and p16 signaling pathways and suppressing oxidative stress.
Humans
;
Mesenchymal Stem Cells/drug effects*
;
Cellular Senescence/drug effects*
;
Chalcone/pharmacology*
;
Oxidative Stress/drug effects*
;
Quinones/pharmacology*
;
Umbilical Cord/cytology*
;
Reactive Oxygen Species/metabolism*
;
Cells, Cultured
;
Cyclin-Dependent Kinase Inhibitor p16/metabolism*
;
Tumor Suppressor Protein p53/metabolism*
;
Membrane Potential, Mitochondrial
;
Cell Proliferation
9.Exploiting targeted degradation of cyclins and cyclin-dependent kinases for cancer therapeutics: a review.
Suya ZHENG ; Ye CHEN ; Zhipeng ZHU ; Nan LI ; Chunyu HE ; H Phillip KOEFFLER ; Xin HAN ; Qichun WEI ; Liang XU
Journal of Zhejiang University. Science. B 2025;26(8):713-739
Cancer is characterized by abnormal cell proliferation. Cyclins and cyclin-dependent kinases (CDKs) have been recognized as essential regulators of the intricate cell cycle, orchestrating DNA replication and transcription, RNA splicing, and protein synthesis. Dysregulation of the CDK pathway is prevalent in the development and progression of human cancers, rendering cyclins and CDKs attractive therapeutic targets. Several CDK4/6 inhibitors have demonstrated promising anti-cancer efficacy and have been successfully translated into clinical use, fueling the development of CDK-targeted therapies. With this enthusiasm for finding novel CDK-targeting anti-cancer agents, there have also been exciting advances in the field of targeted protein degradation through innovative strategies, such as using proteolysis-targeting chimera, heat shock protein 90 (HSP90)-mediated targeting chimera, hydrophobic tag-based protein degradation, and molecular glue. With a focus on the translational potential of cyclin- and CDK-targeting strategies in cancer, this review presents the fundamental roles of cyclins and CDKs in cancer. Furthermore, it summarizes current strategies for the proteasome-dependent targeted degradation of cyclins and CDKs, detailing the underlying mechanisms of action for each approach. A comprehensive overview of the structure and activity of existing CDK degraders is also provided. By examining the structure‒activity relationships, target profiles, and biological effects of reported cyclin/CDK degraders, this review provides a valuable reference for both CDK pathway-targeted biomedical research and cancer therapeutics.
Humans
;
Neoplasms/metabolism*
;
Cyclin-Dependent Kinases/antagonists & inhibitors*
;
Cyclins/metabolism*
;
Proteolysis
;
Antineoplastic Agents/pharmacology*
;
Molecular Targeted Therapy
;
Proteasome Endopeptidase Complex/metabolism*
;
Animals
10.High expression of CDKN3 promotes migration and invasion of gastric cancer cells by regulating the p53/NF-κB signaling pathway and inhibiting cell apoptosis.
Yi ZHANG ; Yu SHEN ; Zhiqiang WAN ; Song TAO ; Yakui LIU ; Shuanhu WANG
Journal of Southern Medical University 2025;45(4):853-861
OBJECTIVES:
To investigate the expression of CDKN3 in gastric cancer and its impact on prognosis of gastric cancer patients.
METHODS:
We analyzed CDKN3 expression in clinical specimens from 114 gastric cancer patients and assessed its association with 5-year postoperative survival of the patients. GO and KEGG enrichment analyses were used to predict the biological function and possible mechanism of CDKN3. The effects of lentivirus-mediated CDKN3 knockdown on biological behaviors of gastric cancer cells were evaluated using Transwell assay, CCK-8 assay, TUNEL staining, flow cytometry, and Western blotting.
RESULTS:
CDKN3 expression was significantly higher in gastric cancer tissues than in the adjacent tissues with significant correlations with CEA level, CA19-9 level, and T and N staging (<i>Pi><0.05). High CDKN3 expression was an independent risk factor affecting 5-year postoperative survival of the patients and predictive for long-term prognosis (<i>Pi><0.01). Enrichment analyses suggested a probable association of CDKN3 with apoptosis. In MGC-803 cells, CDKN3 knockdown significantly lowered migration and invasion capacities of the cells, while CDKN3 overexpression produced the opposite effects. TUNEL staining revealed a significantly lower level of cell apoptosis in gastric cancer tissues than in adjacent tissues (<i>Pi><0.01). CDKN3 knockdown obviously inhibited proliferation and increased apoptosis of MGC-803 cells. CDKN3 overexpression down-regulated the expressions of p53, p21 and Bax and up-regulated the expressions of p-p65 and Bcl-2.
CONCLUSIONS
CDKN3 is highly expressed in gastric cancer tissues and affects patient prognosis. CDKN3 overexpression promotes proliferation, invasion and migration and suppressed apoptosis of gastric cancer cells possibly through the p53/NF-κB signaling pathway.
Humans
;
Stomach Neoplasms/pathology*
;
Apoptosis
;
Signal Transduction
;
Tumor Suppressor Protein p53/metabolism*
;
Cell Movement
;
Cell Line, Tumor
;
NF-kappa B/metabolism*
;
Prognosis
;
Cyclin-Dependent Kinase Inhibitor Proteins/metabolism*
;
Cell Proliferation
;
Neoplasm Invasiveness
;
Male
;
Female
;
Dual-Specificity Phosphatases


Result Analysis
Print
Save
E-mail