1
|
Niebora J, Woźniak S, Domagała D, Data K, Farzaneh M, Zehtabi M, Dari MAG, Pour FK, Bryja A, Kulus M, Mozdziak P, Dzięgiel P, Kempisty B. The role of ncRNAs and exosomes in the development and progression of endometrial cancer. Front Oncol 2024; 14:1418005. [PMID: 39188680 PMCID: PMC11345653 DOI: 10.3389/fonc.2024.1418005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 08/28/2024] Open
Abstract
Endometrial cancer (EC) is one of the most common gynecologic cancers. In recent years, research has focused on the genetic characteristics of the tumors to detail their prognosis and tailor therapy. In the case of EC, genetic mutations have been shown to underlie their formation. It is very important to know the mechanisms of EC formation related to mutations induced by estrogen, among other things. Noncoding RNAs (ncRNAs), composed of nucleotide transcripts with very low protein-coding capacity, are proving to be important. Their expression patterns in many malignancies can inhibit tumor formation and progression. They also regulate protein coding at the epigenetic, transcriptional, and posttranscriptional levels. MicroRNAs (miRNAs), several varieties of which are associated with normal endometrium as well as its tumor, also play a particularly important role in gene expression. MiRNAs and long noncoding RNAs (lncRNAs) affect many pathways in EC tissues and play important roles in cancer development, invasion, and metastasis, as well as resistance to anticancer drugs through mechanisms such as suppression of apoptosis and progression of cancer stem cells. It is also worth noting that miRNAs are highly precise, sensitive, and robust, making them potential markers for diagnosing gynecologic cancers and their progression. Unfortunately, as the incidence of EC increases, treatment becomes challenging and is limited to invasive tools. The prospect of using microRNAs as potential candidates for diagnostic and therapeutic use in EC seems promising. Exosomes are extracellular vesicles that are released from many types of cells, including cancer cells. They contain proteins, DNA, and various types of RNA, such as miRNAs. The noncoding RNA components of exosomes vary widely, depending on the physiology of the tumor tissue and the cells from which they originate. Exosomes contain both DNA and RNA and have communication functions between cells. Exosomal miRNAs mediate communication between EC cells, tumor-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs) and play a key role in tumor cell proliferation and tumor microenvironment formation. Oncogenes carried by tumor exosomes induce malignant transformation of target cells. During the synthesis of exosomes, various factors, such as genetic and proteomic data are upregulated. Thus, they are considered an interesting therapeutic target for the diagnosis and prognosis of endometrial cancer by analyzing biomarkers contained in exosomes. Expression of miRNAs, particularly miR-15a-5p, was elevated in exosomes derived from the plasma of EC patients. This may suggest the important utility of this biomarker in the diagnosis of EC. In recent years, researchers have become interested in the topic of prognostic markers for EC, as there are still too few identified markers to support the limited treatment of endometrial cancer. Further research into the effects of ncRNAs and exosomes on EC may allow for cancer treatment breakthroughs.
Collapse
Affiliation(s)
- Julia Niebora
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Sławomir Woźniak
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Dominika Domagała
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Data
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Clinical Research Development Unit, Imam Khomeini Hospital, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Zehtabi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahrokh Abouali Gale Dari
- Department of Obstetrics and Gynecology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Khojasteh Pour
- Department of Obstetrics and Gynecology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Artur Bryja
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC, United States
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Bartosz Kempisty
- Division of Anatomy, Department of Human Morphology and Embryology, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Torun, Poland
- Physiology Graduate Program, North Carolina State University, Raleigh, NC, United States
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, Brno, Czechia
| |
Collapse
|
2
|
Žvinys G, Petrosiute A, Zakšauskas A, Zubrienė A, Ščerbavičienė A, Kalnina Z, Čapkauskaitė E, Juozapaitienė V, Mickevičiu̅tė A, Shubin K, Grincevičienė Š, Raišys S, Tars K, Matulienė J, Matulis D. High-Affinity NIR-Fluorescent Inhibitors for Tumor Imaging via Carbonic Anhydrase IX. Bioconjug Chem 2024; 35:790-803. [PMID: 38750635 PMCID: PMC11191402 DOI: 10.1021/acs.bioconjchem.4c00144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 06/21/2024]
Abstract
Tumor imaging and delivery of therapeutic agents may be achieved by designing high-affinity and high-selectivity compounds recognizing a tumor cell-expressing biomarker, such as carbonic anhydrase IX (CA IX). The CAIX, overexpressed in many hypoxic solid tumors, helps adjust to the energy requirements of the hypoxic environment, reduces intracellular acidification, and participates in the metastatic invasion of adjacent tissues. Here, we designed a series of sulfonamide compounds bearing CAIX-recognizing, high-affinity, and high-selectivity groups conjugated via a PEG linker to near-infrared (NIR) fluorescent probes used in the clinic for optically guided cancer surgery. We determined compound affinities for CAIX and other 11 catalytically active CA isozymes by the thermal shift assay and showed that the affinity Kd value of CAIX was in the subnanomolar range, hundred to thousand-fold higher than those of other CA isozymes. Similar affinities were also observed for CAIX expressed on the cancer cell surface in live HeLa cell cultures, as determined by the competition assay. The NIR-fluorescent compounds showed excellent properties in visualizing CAIX-positive tumors but not CAIX-negative knockout tumors in a nude mice xenograft model. These compounds would therefore be helpful in optically guided cancer surgery and could potentially be developed for anticancer treatment by radiotherapy.
Collapse
Affiliation(s)
- Gediminas Žvinys
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Agne Petrosiute
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Audrius Zakšauskas
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Asta Zubrienė
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Alvilė Ščerbavičienė
- Department
of Biological Models, Institute of Biochemistry, Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Zane Kalnina
- Latvian
Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| | - Edita Čapkauskaitė
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Vaida Juozapaitienė
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Aurelija Mickevičiu̅tė
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Kirill Shubin
- Latvian
Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Švitrigailė Grincevičienė
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Steponas Raišys
- Institute
of Photonics and Nanotechnology, National Center for Physical Sciences
and Technology, Vilnius University, Saulėtekio 3, Vilnius LT-10257, Lithuania
| | - Kaspars Tars
- Latvian
Biomedical Research and Study Centre, Ratsupites 1 k-1, Riga LV-1067, Latvia
| | - Jurgita Matulienė
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| | - Daumantas Matulis
- Department
of Biothermodynamics and Drug Design, Institute of Biotechnology,
Life Sciences Center, Vilnius University, Saulėtekio 7, Vilnius LT-10257, Lithuania
| |
Collapse
|
3
|
Xiang N, Chen T, Zhao X, Zhao M. In vitro assessment of roles of PPP1R14B in cervical and endometrial cancer. Tissue Cell 2022; 77:101845. [PMID: 35679681 DOI: 10.1016/j.tice.2022.101845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 11/19/2022]
Abstract
Cervical and endometrial cancers are common gynecologic cancers. Protein phosphatase 1 regulatory subunit 14B (PPP1R14B) is aberrantly expressed in several tumors, while its functions in cervical and endometrial cancers remain largely uncertain. The differentially expression of PPP1R14B in cervical and endometrial cancers was predicted by GEPIA2 and Human Protein Atlas databases. The diagnostic value was analyzed by AUC curve. The association between PPP1R14B expression and overall survival was predicted using Kaplan-Meier Plotter database. The function of PPP1R14B was investigated according to in vitro assessment. PPP1R14B and phosphorylation level of Akt were analyzed through western blotting. Cell proliferation was investigated by CCK-8 and EdU staining assays. Cell apoptosis was evaluated via TUNEL staining and caspase-3 activity assays. PPP1R14B level was upregulated in cervical and endometrial cancers, and it was associated with diagnosis and worse prognosis. PPP1R14B silencing constrained cell proliferation and promoted cell death in cervical and endometrial cancers cells. PPP1R14B knockdown suppressed activation of the Akt pathway. Re-activation of the Akt signaling reversed the anti-proliferative and cell death-promoting roles of PP1R14B knockdown in cervical and endometrial cancers cells. In conclusion, PPP1R14B knockdown represses cell proliferation and facilitates cell death by inhibiting the activation of the Akt signaling in cervical and endometrial cancers.
Collapse
Affiliation(s)
- Nan Xiang
- Department of Obstetrics and Gynecology, Shandong Provincial Third Hospital, Jinan 250031, China
| | - Tao Chen
- Department of Clinical Laboratory, Jinan Maternity and Child Care Hospital, Jinan 250001, China
| | - Xiaoli Zhao
- Department of Obstetrics, The Third People's Hospital of Jinan, Jinan 250132, China
| | - Min Zhao
- Department of Obstetrics and Gynecology, Shandong Provincial Third Hospital, Jinan 250031, China.
| |
Collapse
|
4
|
Helderman RFCPA, Restrepo MT, Rodermond HM, van Bochove GGW, Löke DR, Franken NAP, Kok HP, Tanis PJ, Crezee J, Oei AL. Non-Invasive Imaging and Scoring of Peritoneal Metastases in Small Preclinical Animal Models Using Ultrasound: A Preliminary Trial. Biomedicines 2022; 10:biomedicines10071610. [PMID: 35884917 PMCID: PMC9313051 DOI: 10.3390/biomedicines10071610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background: The peritoneum is a common site for the formation of metastases originating from several gastrointestinal and gynecological malignancies. A representative preclinical model to thoroughly explore the pathophysiological mechanisms and to study new treatment strategies is important. A major challenge for such models is defining and quantifying the (total) tumor burden in the peritoneal cavity prior to treatment, since it is preferable to use non-invasive methods. We evaluated ultrasound as a simple and easy-to-handle imaging method for this purpose. Methods: Peritoneal metastases were established in six WAG/Rij rats through i.p. injections of the colon carcinoma cell line CC-531. Using ultrasound, the location, number and size of intraperitoneal tumor nodules were determined by two independent observers. Tumor outgrowth was followed using ultrasound until the peritoneal cancer index (PCI) was ≥8. Interobserver variability and ex vivo correlation were assessed. Results: Visible peritoneal tumor nodules were formed in six WAG/Rij rats within 2–4 weeks after cell injection. In most animals, tumor nodules reached a size of 4–6 mm within 3–4 weeks, with total PCI scores ranging from 10–20. The predicted PCI scores using ultrasound ranged from 11–19 and from 8–18, for observer 1 and 2, respectively, which was quite similar to the ex vivo scores. Conclusions: Ultrasound is a reliable non-invasive method to detect intraperitoneal tumor nodules and quantify tumor outgrowth in a rat model.
Collapse
Affiliation(s)
- Roxan F. C. P. A. Helderman
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.F.C.P.A.H.); (H.M.R.); (G.G.W.v.B.); (D.R.L.); (N.A.P.F.); (H.P.K.); (J.C.)
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
| | - Mauricio Tobón Restrepo
- Division of Diagnostic Imaging, Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands;
| | - Hans M. Rodermond
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.F.C.P.A.H.); (H.M.R.); (G.G.W.v.B.); (D.R.L.); (N.A.P.F.); (H.P.K.); (J.C.)
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
| | - Gregor G. W. van Bochove
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.F.C.P.A.H.); (H.M.R.); (G.G.W.v.B.); (D.R.L.); (N.A.P.F.); (H.P.K.); (J.C.)
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
| | - Daan R. Löke
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.F.C.P.A.H.); (H.M.R.); (G.G.W.v.B.); (D.R.L.); (N.A.P.F.); (H.P.K.); (J.C.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
| | - Nicolaas A. P. Franken
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.F.C.P.A.H.); (H.M.R.); (G.G.W.v.B.); (D.R.L.); (N.A.P.F.); (H.P.K.); (J.C.)
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
| | - H. Petra Kok
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.F.C.P.A.H.); (H.M.R.); (G.G.W.v.B.); (D.R.L.); (N.A.P.F.); (H.P.K.); (J.C.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
| | - Pieter J. Tanis
- Department of Surgery, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands;
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Johannes Crezee
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.F.C.P.A.H.); (H.M.R.); (G.G.W.v.B.); (D.R.L.); (N.A.P.F.); (H.P.K.); (J.C.)
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
| | - Arlene L. Oei
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (R.F.C.P.A.H.); (H.M.R.); (G.G.W.v.B.); (D.R.L.); (N.A.P.F.); (H.P.K.); (J.C.)
- Center for Experimental and Molecular Medicine (CEMM), Laboratory for Experimental Oncology and Radiobiology (LEXOR), 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology, 1105 AZ Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
5
|
Espedal H, Berg HF, Fonnes T, Fasmer KE, Krakstad C, Haldorsen IS. Feasibility and utility of MRI and dynamic 18F-FDG-PET in an orthotopic organoid-based patient-derived mouse model of endometrial cancer. J Transl Med 2021; 19:406. [PMID: 34565386 PMCID: PMC8474962 DOI: 10.1186/s12967-021-03086-9] [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: 04/29/2021] [Accepted: 09/19/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Pelvic magnetic resonance imaging (MRI) and whole-body positron emission tomography-computed tomography (PET-CT) play an important role at primary diagnostic work-up and in detecting recurrent disease in endometrial cancer (EC) patients, however the preclinical use of these imaging methods is currently limited. We demonstrate the feasibility and utility of MRI and dynamic 18F-fluorodeoxyglucose (FDG)-PET imaging for monitoring tumor progression and assessing chemotherapy response in an orthotopic organoid-based patient-derived xenograft (O-PDX) mouse model of EC. METHODS 18 O-PDX mice (grade 3 endometrioid EC, stage IIIC1), selectively underwent weekly T2-weighted MRI (total scans = 32), diffusion-weighted MRI (DWI) (total scans = 9) and dynamic 18F-FDG-PET (total scans = 26) during tumor progression. MRI tumor volumes (vMRI), tumor apparent diffusion coefficient values (ADCmean) and metabolic tumor parameters from 18F-FDG-PET including maximum and mean standard uptake values (SUVmax/SUVmean), metabolic tumor volume (MTV), total lesion glycolysis (TLG) and metabolic rate of 18F-FDG (MRFDG) were calculated. Further, nine mice were included in a chemotherapy treatment study (treatment; n = 5, controls; n = 4) and tumor ADCmean-values were compared to changes in vMRI and cellular density from histology at endpoint. A Mann-Whitney test was used to evaluate differences between groups. RESULTS Tumors with large tumor volumes (vMRI) had higher metabolic activity (MTV and TLG) in a clear linear relationship (r2 = 0.92 and 0.89, respectively). Non-invasive calculation of MRFDG from dynamic 18F-FDG-PET (mean MRFDG = 0.39 μmol/min) was feasible using an image-derived input function. Treated mice had higher tumor ADCmean (p = 0.03), lower vMRI (p = 0.03) and tumor cellular density (p = 0.02) than non-treated mice, all indicating treatment response. CONCLUSION Preclinical imaging mirroring clinical imaging methods in EC is highly feasible for monitoring tumor progression and treatment response in the present orthotopic organoid mouse model.
Collapse
Affiliation(s)
- Heidi Espedal
- Department of Clinical Medicine, University of Bergen, 5021, Bergen, Norway.
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, 5021, Bergen, Norway.
| | - Hege F Berg
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5021, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Tina Fonnes
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5021, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Kristine E Fasmer
- Department of Clinical Medicine, University of Bergen, 5021, Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Camilla Krakstad
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5021, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5021, Bergen, Norway
| | - Ingfrid S Haldorsen
- Department of Clinical Medicine, University of Bergen, 5021, Bergen, Norway
- Mohn Medical Imaging and Visualization Centre, Department of Radiology, Haukeland University Hospital, 5021, Bergen, Norway
| |
Collapse
|
6
|
New Insights into Breast and Endometrial Cancers. Cancers (Basel) 2020; 12:cancers12092595. [PMID: 32932889 PMCID: PMC7563714 DOI: 10.3390/cancers12092595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/08/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
|
7
|
He S, Fang X, Xia X, Hou T, Zhang T. Targeting CDK9: A novel biomarker in the treatment of endometrial cancer. Oncol Rep 2020; 44:1929-1938. [PMID: 32901849 PMCID: PMC7551504 DOI: 10.3892/or.2020.7746] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/21/2020] [Indexed: 12/18/2022] Open
Abstract
Endometrial cancer is one of the three major malignant tumors of the female reproductive system. Although cyclin-dependent kinase 9 (CDK9) has a definitive pathogenic role in various types of cancer, little is known concerning its function in endometrial cancer. Our study was conducted to evaluate the expression and therapeutic potential of CDK9 in endometrial cancer. CDK9 expression was determined by immunohistochemistry in endometrial cancer tissues constructed with paired primary, metastatic, and recurrent tumor tissues from 32 endometrial cancer patients. Small interfering RNA (siRNA) and inhibitors of CDK9 were used to evaluate the effect of CDK9 inhibition on the anti-apoptotic activity and proliferation in endometrial cancer cells. Colony formation assay and wound-healing assays were adopted to assess clonal formation and migratory capacity. The results of the immunohistochemistry demonstrated that CDK9 was highly expressed in the human endometrial cancer cell lines; moreover, it was elevated in metastatic and recurrent endometrial tumor tissue compared when compared with that in patient-matched primary endometrial tumor tissue. Knockdown of CDK9 with siRNA and inhibition of CDK9 activity with the inhibitor suppressed cell proliferation and promoted apoptosis in endometrial cancer. In conclusion, our results provide evidence that CDK9 may be a potential prognostic biomarker and a promising therapeutic target for the treatment of endometrial cancer in the future.
Collapse
Affiliation(s)
- Shasha He
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Xiaoling Fang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Xiaomeng Xia
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Tao Hou
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| | - Tingting Zhang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410000, P.R. China
| |
Collapse
|