1
|
Chen JLY, Pan CK, Lin LC, Tsai CY, Kuo CY, Huang YS, Lin YL. Therapeutic efficacy of cyclin-dependent kinase inhibition in combination with ionizing radiation for lung cancer. Int J Radiat Biol 2023; 99:1257-1266. [PMID: 36598432 DOI: 10.1080/09553002.2023.2161658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 11/24/2022] [Accepted: 12/13/2022] [Indexed: 01/05/2023]
Abstract
PURPOSE To evaluate the therapeutic efficacy of cyclin-dependent kinase (CDK) inhibition in combination with ionizing radiation for lung cancer. MATERIALS AND METHODS Human lung adenocarcinoma (A549) and squamous cell carcinoma (H520) cells were used to evaluate the therapeutic efficacy of CDK inhibition in combination with ionizing radiation in vitro using colony formation assay, γH2AX immunofluorescence staining, western blotting, and cell cycle phase analysis. We also performed in vivo evaluations of ectopic tumor growth. RESULTS In vitro pretreatment with the CDK inhibitor, seliciclib, before irradiation significantly decreased the survival of A549 and H520 cells in a dose-dependent manner. Although CDK inhibition alone did not increase the intensity of γH2AX foci, its combination with ionizing radiation increased DNA double-strand breaks, as shown by γH2AX immunofluorescence staining and western blotting. The combination of CDK inhibition and ionizing radiation-induced G2/M arrest and increased apoptosis, as evidenced by the increased proportion of cells in G2/M arrest, subG1 apoptotic population, and expression of apoptotic markers (cleaved PARP-1 and cleaved caspase-3). Mechanistic studies showed reduced expression of cyclin A with combined treatment, indicating cell cycle shifting effects. An in vivo xenograft model showed that the combination of CDK inhibition and ionizing radiation delayed xenograft tumor growth, and increased the proportion of cleaved PARP-1- and cleaved caspase-3-positive cells, compared to either treatment alone. CONCLUSIONS We provide preclinical tumoricidal evidence that the combination of CDK inhibition and ionizing radiation is an efficacious treatment for lung cancer.
Collapse
Affiliation(s)
- Jenny Ling-Yu Chen
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Radiology, National Taiwan University College of Medicine, Taipei, Taiwan
- National Taiwan University Cancer Center, Taipei, Taiwan
| | - Chun-Kai Pan
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Li-Cheng Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Yi Tsai
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Ying Kuo
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Sen Huang
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Li Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
2
|
Chitsike L, Duerksen-Hughes PJ. Targeted Therapy as a Potential De-Escalation Strategy in Locally Advanced HPV-Associated Oropharyngeal Cancer: A Literature Review. Front Oncol 2021; 11:730412. [PMID: 34490123 PMCID: PMC8418093 DOI: 10.3389/fonc.2021.730412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022] Open
Abstract
The treatment landscape of locally advanced HPV-oropharyngeal squamous cell carcinoma (OPSCC) is undergoing transformation. This is because the high cures rates observed in OPSCC are paired with severe treatment-related, long-term toxicities. These significant adverse effects have led some to conclude that the current standard of care is over-treating patients, and that de-intensifying the regimens may achieve comparable survival outcomes with lower toxicities. Consequently, several de-escalation approaches involving locally advanced OPSCC are underway. These include the reduction of dosage and volume of intensive cytotoxic regimens, as well as elimination of invasive surgical procedures. Such de-intensifying treatments have the potential to achieve efficacy and concurrently alleviate morbidity. Targeted therapies, given their overall safer toxicity profiles, also make excellent candidates for de-escalation, either alone or alongside standard treatments. However, their role in these endeavors is currently limited, because few targeted therapies are currently in clinical use for head and neck cancers. Unfortunately, cetuximab, the only FDA-approved targeted therapy, has shown inferior outcomes when paired with radiation as compared to cisplatin, the standard radio-sensitizer, in recent de-escalation trials. These findings indicate the need for a better understanding of OPSCC biology in the design of rational therapeutic strategies and the development of novel, OPSCC-targeted therapies that are safe and can improve the therapeutic index of standard therapies. In this review, we summarize ongoing research on mechanism-based inhibitors in OPSCC, beginning with the salient molecular features that modulate tumorigenic processes and response, then exploring pharmacological inhibition and pre-clinical validation studies of candidate targeted agents, and finally, summarizing the progression of those candidates in the clinic.
Collapse
|
3
|
Kuchur OA, Kuzmina DO, Dukhinova MS, Shtil AA. The p53 Protein Family in the Response of Tumor Cells to Ionizing Radiation: Problem Development. Acta Naturae 2021; 13:65-76. [PMID: 34707898 PMCID: PMC8526179 DOI: 10.32607/actanaturae.11247] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 12/24/2020] [Indexed: 12/05/2022] Open
Abstract
Survival mechanisms are activated in tumor cells in response to therapeutic ionizing radiation. This reduces a treatment's effectiveness. The p53, p63, and p73 proteins belonging to the family of proteins that regulate the numerous pathways of intracellular signal transduction play a key role in the development of radioresistance. This review analyzes the p53-dependent and p53-independent mechanisms involved in overcoming the resistance of tumor cells to radiation exposure.
Collapse
Affiliation(s)
- O. A. Kuchur
- ITMO University, Saint-Petersburg, 191002 Russia
| | | | | | - A. A. Shtil
- ITMO University, Saint-Petersburg, 191002 Russia
- Blokhin National Medical Research Center of Oncology, Moscow, 115478 Russia
| |
Collapse
|
4
|
Riess C, Irmscher N, Salewski I, Strüder D, Classen CF, Große-Thie C, Junghanss C, Maletzki C. Cyclin-dependent kinase inhibitors in head and neck cancer and glioblastoma-backbone or add-on in immune-oncology? Cancer Metastasis Rev 2021; 40:153-171. [PMID: 33161487 PMCID: PMC7897202 DOI: 10.1007/s10555-020-09940-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/26/2020] [Indexed: 12/11/2022]
Abstract
Cyclin-dependent kinases (CDK) control the cell cycle and play a crucial role in oncogenesis. Pharmacologic inhibition of CDK has contributed to the recent clinical approval of dual CDK4/6 inhibitors for the treatment of breast and small cell lung cancer. While the anticancer cell effects of CDK inhibitors are well-established, preclinical and early clinical studies describe additional mechanisms of action such as chemo- and radiosensitization or immune stimulation. The latter offers great potential to incorporate CDK inhibitors in immune-based treatments. However, dosing schedules and accurate timing of each combination partner need to be respected to prevent immune escape and resistance. In this review, we provide a detailed summary of CDK inhibitors in the two solid cancer types head and neck cancer and glioblastoma multiforme; it describes the molecular mechanisms of response vs. resistance and covers strategies to avoid resistance by the combination of immunotherapy or targeted therapy.
Collapse
Affiliation(s)
- Christin Riess
- Department of Medicine, Clinic III - Hematology, Oncology and Palliative Care, Rostock University Medical Center, Rostock, Germany
- University Children's and Adolescents' Hospital, Rostock University Medical Center, Rostock, Germany
| | - Nina Irmscher
- Department of Medicine, Clinic III - Hematology, Oncology and Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - Inken Salewski
- Department of Medicine, Clinic III - Hematology, Oncology and Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - Daniel Strüder
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery "Otto Körner", Rostock University Medical Center, Rostock, Germany
| | - Carl-Friedrich Classen
- University Children's and Adolescents' Hospital, Rostock University Medical Center, Rostock, Germany
| | - Christina Große-Thie
- Department of Medicine, Clinic III - Hematology, Oncology and Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - Christian Junghanss
- Department of Medicine, Clinic III - Hematology, Oncology and Palliative Care, Rostock University Medical Center, Rostock, Germany
| | - Claudia Maletzki
- Department of Medicine, Clinic III - Hematology, Oncology and Palliative Care, Rostock University Medical Center, Rostock, Germany.
| |
Collapse
|
5
|
Garcia-Oliveira P, Otero P, Pereira AG, Chamorro F, Carpena M, Echave J, Fraga-Corral M, Simal-Gandara J, Prieto MA. Status and Challenges of Plant-Anticancer Compounds in Cancer Treatment. Pharmaceuticals (Basel) 2021; 14:ph14020157. [PMID: 33673021 PMCID: PMC7918405 DOI: 10.3390/ph14020157] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 12/12/2022] Open
Abstract
Nowadays, cancer is one of the deadliest diseases in the world, which has been estimated to cause 9.9 million deaths in 2020. Conventional treatments for cancer commonly involve mono-chemotherapy or a combination of radiotherapy and mono-chemotherapy. However, the negative side effects of these approaches have been extensively reported and have prompted the search of new therapeutic drugs. In this context, scientific community started to look for innovative sources of anticancer compounds in natural sources, including traditional plants. Currently, numerous studies have evaluated the anticancer properties of natural compounds derived from plants, both in vitro and in vivo. In pre-clinical stages, some promising compounds could be mentioned, such as the sulforaphane or different phenolic compounds. On the other hand, some phytochemicals obtained positive results in clinical stages and were further approved for cancer treatment, such as vinca alkaloids or the paclitaxel. Nevertheless, these compounds are not exempt of limitations, such as low solubility, restricted effect on their own, negative side-effects, etc. This review aims to compile the information about the current phytochemicals used for cancer treatment and also promising candidates, main action mechanisms and also reported limitations. In this sense, some strategies to face the limitations have been considered, such as nano-based formulations to improve solubility or chemical modification to reduce toxicity. In conclusion, although more research is still necessary to develop more efficient and safe phytochemical drugs, more of these compounds might be used in future cancer therapies.
Collapse
Affiliation(s)
- Paula Garcia-Oliveira
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolonia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| | - Paz Otero
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Antia Gonzalez Pereira
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolonia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| | - Franklin Chamorro
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Maria Carpena
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Javier Echave
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Maria Fraga-Corral
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolonia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Correspondence: (J.S.-G.); (M.A.P.)
| | - Miguel Angel Prieto
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Correspondence: (J.S.-G.); (M.A.P.)
| |
Collapse
|
6
|
Figueroa-González G, Carrillo-Hernández JF, Perez-Rodriguez I, Cantú de León D, Campos-Parra AD, Martínez-Gutiérrez AD, Coronel-Hernández J, García-Castillo V, López-Camarillo C, Peralta-Zaragoza O, Jacobo-Herrera NJ, Guardado-Estrada M, Pérez-Plasencia C. Negative Regulation of Serine Threonine Kinase 11 (STK11) through miR-100 in Head and Neck Cancer. Genes (Basel) 2020; 11:E1058. [PMID: 32911741 PMCID: PMC7563199 DOI: 10.3390/genes11091058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/18/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Serine Threonine Kinase 11 (STK11), also known as LKB1, is a tumor suppressor gene that regulates several biological processes such as apoptosis, energetic metabolism, proliferation, invasion, and migration. During malignant progression, different types of cancer inhibit STK11 function by mutation or epigenetic inactivation. In Head and Neck Cancer, it is unclear what mechanism is involved in decreasing STK11 levels. Thus, the present work aims to determine whether STK11 expression might be regulated through epigenetic or post-translational mechanisms. METHODS Expression levels and methylation status for STK11 were analyzed in 59 cases of head and neck cancer and 10 healthy tissue counterparts. Afterward, we sought to identify candidate miRNAs exerting post-transcriptional regulation of STK11. Then, we assessed a luciferase gene reporter assay to know if miRNAs directly target STK11 mRNA. The expression levels of the clinical significance of mir-100-3p, -5p, and STK11 in 495 HNC specimens obtained from the TCGA database were further analyzed. Finally, the Kaplan-Meier method was used to estimate the prognostic significance of the miRNAs for Overall Survival, and survival curves were compared through the log-rank test. RESULTS STK11 was under-expressed, and its promoter region was demethylated or partially methylated. miR-17-5p, miR-106a-5p, miR-100-3p, and miR-100-5p could be negative regulators of STK11. Our experimental data suggested evidence that miR-100-3p and -5p were over-expressed in analyzed tumor patient samples. Luciferase gene reporter assay experiments showed that miR-100-3p targets and down-regulates STK11 mRNA directly. With respect to overall survival, STK11 expression level was significant for predicting clinical outcomes. CONCLUSION This is, to our knowledge, the first report of miR-100-3p targeting STK11 in HNC. Together, these findings may support the importance of regulation of STK11 through post-transcriptional regulation in HNC and the possible contribution to the carcinogenesis process in this neoplasia.
Collapse
Affiliation(s)
- Gabriela Figueroa-González
- Unidad Multidisciplinaria de Investigación Experimental Zaragoza (UMIEZ), Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Mexico City 09230, Mexico;
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - José F. Carrillo-Hernández
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Itzel Perez-Rodriguez
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - David Cantú de León
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Alma D. Campos-Parra
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Antonio D. Martínez-Gutiérrez
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Jossimar Coronel-Hernández
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
| | - Verónica García-Castillo
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica del Cáncer, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Edo.Mex, Mexico;
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City 09790, Mexico;
| | - Oscar Peralta-Zaragoza
- Dirección de Infecciones Crónicas y Cáncer, Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca 62100, Morelos, Mexico;
| | - Nadia J. Jacobo-Herrera
- Unidad de Bioquímica, Instituto Nacional de Nutrición y Ciencias Médicas, Salvador Zubirán, Mexico City 14000, Mexico;
| | - Mariano Guardado-Estrada
- Laboratorio de Genética, Licenciatura en Ciencia Forense, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico;
| | - Carlos Pérez-Plasencia
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica, Instituto Nacional de Cancerología, Mexico City 14080, Mexico; (J.F.C.-H.); (I.P.-R.); (D.C.d.L.); (A.D.C.-P.); (A.D.M.-G.); (J.C.-H.)
- Unidad de Investigación Biomédica en Cáncer, Laboratorio de Genómica del Cáncer, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Edo.Mex, Mexico;
| |
Collapse
|
7
|
Oner M, Lin E, Chen MC, Hsu FN, Shazzad Hossain Prince GM, Chiu KY, Teng CLJ, Yang TY, Wang HY, Yue CH, Yu CH, Lai CH, Hsieh JT, Lin H. Future Aspects of CDK5 in Prostate Cancer: From Pathogenesis to Therapeutic Implications. Int J Mol Sci 2019; 20:ijms20163881. [PMID: 31395805 PMCID: PMC6720211 DOI: 10.3390/ijms20163881] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 01/03/2023] Open
Abstract
Cyclin-dependent kinase 5 (CDK5) is a unique member of the cyclin-dependent kinase family. CDK5 is activated by binding with its regulatory proteins, mainly p35, and its activation is essential in the development of the central nervous system (CNS) and neurodegeneration. Recently, it has been reported that CDK5 plays important roles in regulating various biological and pathological processes, including cancer progression. Concerning prostate cancer, the androgen receptor (AR) is majorly involved in tumorigenesis, while CDK5 can phosphorylate AR and promotes the proliferation of prostate cancer cells. Clinical evidence has also shown that the level of CDK5 is associated with the progression of prostate cancer. Interestingly, inhibition of CDK5 prevents prostate cancer cell growth, while drug-triggered CDK5 hyperactivation leads to apoptosis. The blocking of CDK5 activity by its small interfering RNAs (siRNA) or Roscovitine, a pan-CDK inhibitor, reduces the cellular AR protein level and triggers the death of prostate cancer cells. Thus, CDK5 plays a crucial role in the growth of prostate cancer cells, and AR regulation is one of the important pathways. In this review paper, we summarize the significant studies on CDK5-mediated regulation of prostate cancer cells. We propose that the CDK5–p35 complex might be an outstanding candidate as a diagnostic marker and potential target for prostate cancer treatment in the near future.
Collapse
Affiliation(s)
- Muhammet Oner
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | - Eugene Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Urology, Chang Bing Show Chwan Memorial Hospital, Changhua 505, Taiwan
| | - Mei-Chih Chen
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Fu-Ning Hsu
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | | | - Kun-Yuan Chiu
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Chieh-Lin Jerry Teng
- Division of Hematology/Medical Oncology, Department of Internal, Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Tsung-Ying Yang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Hsin-Yi Wang
- Department of Nuclear Medicine, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Chia-Herng Yue
- Department of Surgery, Tung's Taichung Metro Harbor Hospital, Taichung 435, Taiwan
| | - Ching-Han Yu
- Department of Physiology, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Chang Gung Medical University, Taoyuan 33302, Taiwan
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan.
- Program in Translational Medicine and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
8
|
Pan C, Issaeva N, Yarbrough WG. HPV-driven oropharyngeal cancer: current knowledge of molecular biology and mechanisms of carcinogenesis. CANCERS OF THE HEAD & NECK 2018; 3:12. [PMID: 31093365 PMCID: PMC6460765 DOI: 10.1186/s41199-018-0039-3] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/09/2018] [Indexed: 12/21/2022]
Abstract
Understanding of oropharyngeal squamous cell carcinoma has significantly progressed over the last decades, and the concept that this disease can be subdivided into two distinct entities based on human papilloma virus (HPV) status has gained acceptance. To combat the constantly growing epidemic of HPV+ oropharyngeal cancer, further investigation and characterization the unique features of the disease, along with the development and implementation of new, targeted therapies, is crucial. In this review, we summarize the etiology, pathogenesis, diagnosis, treatment, and molecular characteristics of HPV-associated oropharyngeal squamous cell carcinoma.
Collapse
Affiliation(s)
- Cassie Pan
- 1Department of Surgery, Division of Otolaryngology, Yale University, New Haven, CT USA
| | - Natalia Issaeva
- 2Department of Otolaryngology/Head and Neck Surgery; Lineberger Cancer Center, University of North Carolina at Chapel Hill, 170 Manning Drive, Campus Box 7070, Chapel Hill, NC 27599 USA
| | - Wendell G Yarbrough
- 2Department of Otolaryngology/Head and Neck Surgery; Lineberger Cancer Center, University of North Carolina at Chapel Hill, 170 Manning Drive, Campus Box 7070, Chapel Hill, NC 27599 USA
| |
Collapse
|
9
|
Chen Y, Hou Y, Ge R, Han J, Xu J, Chen J, Wang H. Protective effect of roscovitine against rotenone-induced parkinsonism. Restor Neurol Neurosci 2018; 36:629-638. [PMID: 30056439 DOI: 10.3233/rnn-180817] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
BACKGROUND Protective effect of roscovitine and deregulation of the p-RB/E2F1 have not been well studied in PD models generated by repeated oral administration of rotenone. OBJECTIVE These experiments evaluated the effects of repeated oral gavage of rotenone on the activation of p-RB/E2F1 and the effects of roscovitine on the regulation of dopaminergic neuronal injury and the behavior of PD in mice. METHODS Using 2.5% carboxymethylcellulose and 1.25% chloroform as a vehicle solution, rotenone (30 mg/kg) was administered via oral gavage once daily for 30 days in C57 mice. Behavioral profiles (pole test and traction test) were assessed in these PD models, and oxidative stress levels were evaluated in the midbrain. The immunoreactivities of TH, α-synuclein (α-syn), p-RB, E2F1 and cleaved caspase-3 in the substantia nigra were examined with a laser confocal microscope. Pharmacological inhibition of cyclin-dependent kinase with roscovitine was achieved by intraperitoneal (IP) injection at a dose of 50 mg/kg daily. RESULTS All rotenone-administered C57 mice showed the typical behavioral features of PD: stiffness, bradykinesia, or hypokinesia. Behavioral testing with the pole test and traction test indicated that the rotenone group, but not the vehicle group, was affected. Spectrophotometric analysis demonstrated that glutathione (GSH) and superoxide dismutase (SOD) activity was decreased, and the generation of malondialdehyde (MDA) was elevated in the midbrain of the rotenone-treated group. After oral administration of rotenone, a loss of nigral tyrosine hydroxylase (TH)-positive neurons was observed. The immune response of α-syn was enhanced in the cytoplasm of dopaminergic neurons from the rotenone-induced neurotoxicity. Rb phosphorylation at serine 780, which affected Rb binding to E2F, was induced after rotenone treatment. The activation of E2F1, which is involved in the regulation of the cell cycle, was also induced from chronic exposure to rotenone. Moreover, administration of the cell cycle inhibitor roscovitine protected against rotenone-induced nigral dopaminergic neuronal injury and inhibited cleaved caspase-3 activation. Roscovitine also markedly ameliorated the behavior of PD mice. CONCLUSIONS Mouse models of Parkinson's disease were established by oral rotenone administration and reproduced some of the features of dopaminergic neuronal degeneration. Roscovitine protects against rotenone-induced parkinsonism.
Collapse
Affiliation(s)
- Yan Chen
- Department of Gastroenterology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Yiwei Hou
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Ruli Ge
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Jianmei Han
- Department of Neurology, Yangxin County People's Hospital, Shandong Province, China
| | - Jing Xu
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Jinbo Chen
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| | - Hongcai Wang
- Department of Neurology, Affiliated Hospital of Binzhou Medical University, Shandong Province, China
| |
Collapse
|
10
|
Kahue CN, Jerrell RJ, Parekh A. Expression of human papillomavirus oncoproteins E6 and E7 inhibits invadopodia activity but promotes cell migration in HPV-positive head and neck squamous cell carcinoma cells. Cancer Rep (Hoboken) 2018; 1:e1125. [PMID: 32721084 PMCID: PMC7941430 DOI: 10.1002/cnr2.1125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/30/2022] Open
Abstract
Background The rapid increase in the incidence of head and neck squamous cell carcinoma (HNSCC) is caused by high‐risk human papillomavirus (HPV) infections. The HPV oncogenes E6 and E7 promote carcinogenesis by disrupting signaling pathways that control survival and proliferation. Although these cancers are often diagnosed with metastases, the mechanisms that regulate their dissemination are unknown. Aims The aim of this study was to determine whether the HPV‐16 E6 and E7 oncogenes affected the invasive and migratory properties of HNSCC cells which promote their spread and metastasis. Methods and results Invasiveness was determined using invadopodia assays which allow for quantitation of extracellular matrix (ECM) degradation by invadopodia which are proteolytic membrane protrusions that facilitate invasion. Using cell lines and genetic manipulations, we found that HPV inhibited invadopodia activity in aggressive cell lines which was mediated by the E6 and E7 oncogenes. Given these findings, we also tested whether HPV caused differences in the migratory ability of HNSCC cells using Transwell assays. In contrast to our invadopodia results, we found no correlation between HPV status and cell migration; however, blocking the expression of the E6 and E7 oncoproteins in a HPV‐positive (HPV+) HNSCC cell line resulted in decreased migration. Conclusions Our data suggest that the E6 and E7 oncoproteins are negative regulators of invadopodia activity but may promote migration in HPV+ HNSCC cells. Despite the need for ECM proteolysis to penetrate most tissues, the unique structure of the head and neck tissues in which these cancers arise may facilitate the spread of migratory cancer cells without significant proteolytic ability.
Collapse
Affiliation(s)
- Charissa N Kahue
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachel J Jerrell
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aron Parekh
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA.,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
11
|
Ozfiliz-Kilbas P, Sarikaya B, Obakan-Yerlikaya P, Coker-Gurkan A, Arisan ED, Temizci B, Palavan-Unsal N. Cyclin-dependent kinase inhibitors, roscovitine and purvalanol, induce apoptosis and autophagy related to unfolded protein response in HeLa cervical cancer cells. Mol Biol Rep 2018; 45:815-828. [PMID: 29978381 DOI: 10.1007/s11033-018-4222-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/25/2018] [Indexed: 12/11/2022]
Abstract
Roscovitine (Rosc) and purvalanol (Pur) are competitive inhibitors of cyclin-dependent kinases (CDKs) by targeting their ATP-binding pockets. Both drugs are shown to be effective to decrease cell viability and dysregulate the ratio of pro- and anti-apoptotic Bcl-2 family members, which finally led to apoptotic cell death in different cancer cell lines in vitro. It was well established that Bcl-2 family members have distinct roles in the regulation of other cellular processes such as endoplasmic reticulum (ER) stress. The induction of ER stress has been shown to play critical role in cell death/survival decision via autophagy or apoptosis. In this study, our aim was to investigate the molecular targets of CDK inhibitors on ER stress mechanism related to distinct cell death types in time-dependent manner in HeLa cervical cancer cells. Our results showed that Rosc and Pur decreased the cell viability, cell growth and colony formation, induced ER stress-mediated autophagy or apoptosis in time-dependent manner. Thus, we conclude that exposure of cells to CDK inhibitors induces unfolded protein response and ER stress leading to autophagy and apoptosis processes in HeLa cervical cancer cells.
Collapse
Affiliation(s)
- Pelin Ozfiliz-Kilbas
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Bahar Sarikaya
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Pinar Obakan-Yerlikaya
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey.
| | - Ajda Coker-Gurkan
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Elif Damla Arisan
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Benan Temizci
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| | - Narcin Palavan-Unsal
- Department of Molecular Biology and Genetics, Istanbul Kultur University, Atakoy Campus, 34156, Istanbul, Turkey
| |
Collapse
|
12
|
Ziemann F, Seltzsam S, Dreffke K, Preising S, Arenz A, Subtil FSB, Rieckmann T, Engenhart-Cabillic R, Dikomey E, Wittig A. Roscovitine strongly enhances the effect of olaparib on radiosensitivity for HPV neg. but not for HPV pos. HNSCC cell lines. Oncotarget 2017; 8:105170-105183. [PMID: 29285242 PMCID: PMC5739629 DOI: 10.18632/oncotarget.22005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 10/04/2017] [Indexed: 02/06/2023] Open
Abstract
At present, advanced stage human Papillomavirus (HPV) negative and positive head and neck squamous cell carcinoma (HNSCC) are treated by intense multimodal therapy that includes radiochemotherapy, which are associated with relevant side effects. Patients with HPV positive tumors possess a far better prognosis than those with HPV negative cancers. Therefore, new therapeutic strategies are needed to improve the outcome especially of the latter one as well as quality of life for all HNSCC patients. Here we tested whether roscovitine, an inhibitor of cyclin-dependent kinases (CDKs), which hereby also blocks homologous recombination (HR), can be used to enhance the radiation sensitivity of HNSCC cell lines. In all five HPV negative and HPV positive cell lines tested, roscovitine caused inhibition of CDK1 and 2. Surprisingly, all HPV positive cell lines were found to be defective in HR. In contrast, HPV negative strains demonstrated efficient HR, which was completely suppressed by roscovitine. In line with this, for HPV negative but not for HPV positive cell lines, treatment with roscovitine resulted in a pronounced enhancement of the radiation-induced G2 arrest as well as a significant increase in radiosensitivity. Due to a defect in HR, all HPV positive cell lines were efficiently radiosensitized by the PARP-1 inhibitor olaparib. In contrast, in HPV negative cell lines a significant radiosensitization by olaparib was only achieved when combined with roscovitine.
Collapse
Affiliation(s)
- Frank Ziemann
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital GieΔen and Marburg, Marburg, Germany
| | - Steve Seltzsam
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital GieΔen and Marburg, Marburg, Germany
| | - Kristin Dreffke
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital GieΔen and Marburg, Marburg, Germany
| | - Stefanie Preising
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital GieΔen and Marburg, Marburg, Germany
| | - Andrea Arenz
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital GieΔen and Marburg, Marburg, Germany
| | - Florentine S B Subtil
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital GieΔen and Marburg, Marburg, Germany
| | - Thorsten Rieckmann
- Laboratory for Radiobiology & Experimental Radiooncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany.,Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Rita Engenhart-Cabillic
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital GieΔen and Marburg, Marburg, Germany
| | - Ekkehard Dikomey
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital GieΔen and Marburg, Marburg, Germany.,Laboratory for Radiobiology & Experimental Radiooncology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Andrea Wittig
- Department of Radiotherapy and Radiooncology, Philipps-University Marburg, University Hospital GieΔen and Marburg, Marburg, Germany
| |
Collapse
|
13
|
Biktasova A, Hajek M, Sewell A, Gary C, Bellinger G, Deshpande HA, Bhatia A, Burtness B, Judson B, Mehra S, Yarbrough WG, Issaeva N. Demethylation Therapy as a Targeted Treatment for Human Papillomavirus-Associated Head and Neck Cancer. Clin Cancer Res 2017; 23:7276-7287. [PMID: 28916527 DOI: 10.1158/1078-0432.ccr-17-1438] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/01/2017] [Accepted: 09/13/2017] [Indexed: 11/16/2022]
Abstract
Purpose: DNA methylation in human papillomavirus-associated (HPV+) head and neck squamous cell carcinoma (HNSCC) may have importance for continuous expression of HPV oncogenes, tumor cell proliferation, and survival. Here, we determined activity of a global DNA-demethylating agent, 5-azacytidine (5-aza), against HPV+ HNSCC in preclinical models and explored it as a targeted therapy in a window trial enrolling patients with HPV+ HNSCC.Experimental Design: Sensitivity of HNSCC cells to 5-aza treatment was determined, and then 5-aza activity was tested in vivo using xenografted tumors in a mouse model. Finally, tumor samples from patients enrolled in a window clinical trial were analyzed to identify activity of 5-aza therapy in patients with HPV+ HNSCC.Results: Clinical trial and experimental data show that 5-aza induced growth inhibition and cell death in HPV+ HNSCC. 5-aza reduced expression of HPV genes, stabilized p53, and induced p53-dependent apoptosis in HNSCC cells and tumors. 5-aza repressed expression and activity of matrix metalloproteinases (MMP) in HPV+ HNSCC, activated IFN response in some HPV+ head and neck cancer cells, and inhibited the ability of HPV+ xenografted tumors to invade mouse blood vessels.Conclusions: 5-aza may provide effective therapy for HPV-associated HNSCC as an alternative or complement to standard cytotoxic therapy. Clin Cancer Res; 23(23); 7276-87. ©2017 AACR.
Collapse
Affiliation(s)
- Asel Biktasova
- Department of Surgery, Division of Otolaryngology, Yale University, New Haven, Connecticut
| | - Michael Hajek
- Department of Surgery, Division of Otolaryngology, Yale University, New Haven, Connecticut
| | - Andrew Sewell
- Department of Surgery, Division of Otolaryngology, Yale University, New Haven, Connecticut
| | - Cyril Gary
- Department of Surgery, Division of Otolaryngology, Yale University, New Haven, Connecticut
| | - Gary Bellinger
- Department of Surgery, Division of Otolaryngology, Yale University, New Haven, Connecticut
| | - Hari A Deshpande
- Department of Medicine, Division of Medical Oncology, Yale University, New Haven, Connecticut
| | - Aarti Bhatia
- Department of Medicine, Division of Medical Oncology, Yale University, New Haven, Connecticut
| | - Barbara Burtness
- Department of Medicine, Division of Medical Oncology, Yale University, New Haven, Connecticut.,Yale Cancer Center, Yale University, New Haven, Connecticut
| | - Benjamin Judson
- Department of Surgery, Division of Otolaryngology, Yale University, New Haven, Connecticut.,Yale Cancer Center, Yale University, New Haven, Connecticut
| | - Saral Mehra
- Department of Surgery, Division of Otolaryngology, Yale University, New Haven, Connecticut
| | - Wendell G Yarbrough
- Department of Surgery, Division of Otolaryngology, Yale University, New Haven, Connecticut. .,Yale Cancer Center, Yale University, New Haven, Connecticut.,Department of Pathology, Yale University, New Haven, Connecticut
| | - Natalia Issaeva
- Department of Surgery, Division of Otolaryngology, Yale University, New Haven, Connecticut. .,Yale Cancer Center, Yale University, New Haven, Connecticut
| |
Collapse
|
14
|
Van Waes C, Musbahi O. Genomics and advances towards precision medicine for head and neck squamous cell carcinoma. Laryngoscope Investig Otolaryngol 2017; 2:310-319. [PMID: 29094075 PMCID: PMC5655563 DOI: 10.1002/lio2.86] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/05/2017] [Indexed: 12/15/2022] Open
Abstract
Objective To provide a review of emerging knowledge from genomics and related basic science, preclinical, and clinical precision medicine studies in head and neck squamous cell carcinoma (HNSCC). Data Sources The Cancer Genome Atlas Network (TCGA) publications, PubMed‐based literature review, and ClinicalTrials.gov. Review Methods TCGA publications, PubMed, and ClinicalTrials.gov were queried for genomics and related basic science, preclinical, and developmental clinical precision medicine studies in HNSCC. Results TCGA reported comprehensive genomic analyses of 279 HNSCC, defining the landscape and frequency of chromosomal copy number alterations, mutations, and expressed genes that contribute to pathogenesis, prognosis, and resistance to therapy. This provides a road map for basic science and preclinical studies to identify key pathways in cancer and cells of the tumor microenvironment affected by these alterations, and candidate targets for new small molecule and biologic therapies. Conclusion Recurrent chromosomal abnormalities, mutations, and expression of genes affecting HNSCC subsets are associated with differences in prognosis, and define molecules, pathways, and deregulated immune responses as candidates for therapy. Activity of molecularly targeted agents appears to be enhanced by rational combinations of these agents and standard therapies targeting the complex alterations that affect multiple pathways and mechanisms in HNSCC. Level of Evidence NA.
Collapse
Affiliation(s)
- Carter Van Waes
- Head and Neck Surgery Branch National Institute on Deafness and Other Communication Disorders Bethesda Maryland U.S.A
| | - Omar Musbahi
- Head and Neck Surgery Branch National Institute on Deafness and Other Communication Disorders Bethesda Maryland U.S.A
| |
Collapse
|
15
|
Wamsley JJ, Gary C, Biktasova A, Hajek M, Bellinger G, Virk R, Issaeva N, Yarbrough WG. Loss of LZAP inactivates p53 and regulates sensitivity of cells to DNA damage in a p53-dependent manner. Oncogenesis 2017; 6:e314. [PMID: 28394357 PMCID: PMC5520489 DOI: 10.1038/oncsis.2017.12] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/22/2016] [Accepted: 02/10/2017] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy and radiation, the two most common cancer therapies, exert their anticancer effects by causing damage to cellular DNA. However, systemic treatment damages DNA not only in cancer, but also in healthy cells, resulting in the progression of serious side effects and limiting efficacy of the treatment. Interestingly, in response to DNA damage, p53 seems to play an opposite role in normal and in the majority of cancer cells-wild-type p53 mediates apoptosis in healthy tissues, attributing to the side effects, whereas mutant p53 often is responsible for acquired cancer resistance to the treatment. Here, we show that leucine zipper-containing ARF-binding protein (LZAP) binds and stabilizes p53. LZAP depletion eliminates p53 protein independently of its mutation status, subsequently protecting wild-type p53 cells from DNA damage-induced cell death, while rendering cells expressing mutant p53 more sensitive to the treatment. In human non-small-cell lung cancer, LZAP levels correlated with p53 levels, suggesting that loss of LZAP may represent a novel mechanism of p53 inactivation in human cancer. Our studies establish LZAP as a p53 regulator and p53-dependent determinative of cell fate in response to DNA damaging treatment.
Collapse
Affiliation(s)
- J J Wamsley
- Division of Otolaryngology, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - C Gary
- Division of Otolaryngology, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - A Biktasova
- Division of Otolaryngology, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - M Hajek
- Division of Otolaryngology, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - G Bellinger
- Division of Otolaryngology, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - R Virk
- Division of Otolaryngology, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
| | - N Issaeva
- Division of Otolaryngology, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - W G Yarbrough
- Division of Otolaryngology, Department of Surgery, Yale University School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|