1
|
Shou S, Maolan A, Zhang D, Jiang X, Liu F, Li Y, Zhang X, Geer E, Pu Z, Hua B, Guo Q, Zhang X, Pang B. Telomeres, telomerase, and cancer: mechanisms, biomarkers, and therapeutics. Exp Hematol Oncol 2025; 14:8. [PMID: 39871386 PMCID: PMC11771031 DOI: 10.1186/s40164-025-00597-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Telomeres and telomerase play crucial roles in the initiation and progression of cancer. As biomarkers, they aid in distinguishing benign from malignant tissues. Despite the promising therapeutic potential of targeting telomeres and telomerase for therapy, translating this concept from the laboratory to the clinic remains challenging. Many candidate drugs remain in the experimental stage, with only a few advancing to clinical trials. This review explores the relationship between telomeres, telomerase, and cancer, synthesizing their roles as biomarkers and reviewing the outcomes of completed trials. We propose that changes in telomere length and telomerase activity can be used to stratify cancer stages. Furthermore, we suggest that differential expression of telomere and telomerase components at the subcellular level holds promise as a biomarker. From a therapeutic standpoint, combining telomerase-targeted therapies with drugs that mitigate the adverse effects of telomerase inhibition may offer a viable strategy.
Collapse
Affiliation(s)
- Songting Shou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ayidana Maolan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Di Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fudong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiyuan Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - En Geer
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenqing Pu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qiujun Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xing Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Bo Pang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
2
|
Gunasekaran P, Han HJ, Choi JH, Ryu EK, Park NY, Bang G, La YK, Park S, Hwang K, Kim HN, Kim MH, Jeon YH, Soung NK, Bang JK. Amphipathic Small Molecule AZT Compound Displays Potent Inhibitory Effects in Cancer Cell Proliferation. Pharmaceutics 2021; 13:pharmaceutics13122071. [PMID: 34959352 PMCID: PMC8704889 DOI: 10.3390/pharmaceutics13122071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 11/24/2022] Open
Abstract
Cancer has been identified as a leading cause of death worldwide, and the increasing number of cancer cases threatens to shorten the average life expectancy of people. Recently, we reported a 3-azido-3-deoxythymidine (AZT)-based amphipathic small molecule, ADG-2e that revealed a notable potency against tumor metastasis. To evaluate the anticancer potential of ADG-2e, we assessed its anticancer potency in vitro and in vivo. Anticancer screening of ADG-2e against cervical cancer cells, HeLa CCL2, and BT549 mammary gland ductal carcinoma showed significant inhibition of cancer cell proliferation. Furthermore, mechanistic investigations revealed that cancer cell death presumably proceeded through an oncosis mechanistic pathway because ADG-2e treated cells showed severe damage on the plasma membrane, a loss of membrane integrity, and leakage of α-tubulin and β-actin. Finally, evaluation of the antitumorigenic potential of ADG-2e in mouse xenograft models revealed that this compound potentially inhibits cancer cell proliferation. Collectively, these findings suggest that ADG-2e can evolve as an anticancer agent, which may represent a model for nucleoside-based small molecule anticancer drug discovery.
Collapse
Affiliation(s)
- Pethaiah Gunasekaran
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Korea; (P.G.); (E.K.R.); (N.Y.P.); (Y.K.L.); (S.P.); (K.H.); (H.N.K.)
- Dandicure Inc., Ochang, Cheongju 28119, Korea
| | - Ho Jin Han
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju 28116, Korea;
| | - Jung hoon Choi
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Cheongju 28119, Korea; (J.h.C.); (G.B.)
| | - Eun Kyoung Ryu
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Korea; (P.G.); (E.K.R.); (N.Y.P.); (Y.K.L.); (S.P.); (K.H.); (H.N.K.)
- Department of Bio-Analytical Science, University of Science & Technology, Daejeon 34113, Korea
| | - Nam Yeong Park
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Korea; (P.G.); (E.K.R.); (N.Y.P.); (Y.K.L.); (S.P.); (K.H.); (H.N.K.)
- Department of Bio-Analytical Science, University of Science & Technology, Daejeon 34113, Korea
| | - Geul Bang
- Biomedical Omics Group, Korea Basic Science Institute, Ochang, Cheongju 28119, Korea; (J.h.C.); (G.B.)
| | - Yeo Kyung La
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Korea; (P.G.); (E.K.R.); (N.Y.P.); (Y.K.L.); (S.P.); (K.H.); (H.N.K.)
| | - Sunghyun Park
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Korea; (P.G.); (E.K.R.); (N.Y.P.); (Y.K.L.); (S.P.); (K.H.); (H.N.K.)
| | - Kyubin Hwang
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Korea; (P.G.); (E.K.R.); (N.Y.P.); (Y.K.L.); (S.P.); (K.H.); (H.N.K.)
| | - Hak Nam Kim
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Korea; (P.G.); (E.K.R.); (N.Y.P.); (Y.K.L.); (S.P.); (K.H.); (H.N.K.)
| | - Mi-Hyun Kim
- Department of Internal Medicine, Pusan National University School of Medicine and Biomedical Research Institute, Pusan National University Hospital, Busan 49241, Korea;
| | - Young Ho Jeon
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Korea
- Correspondence: (Y.H.J.); (N.-K.S.); (J.K.B.)
| | - Nak-Kyun Soung
- Anticancer Agent Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang, Cheongju 28116, Korea;
- Correspondence: (Y.H.J.); (N.-K.S.); (J.K.B.)
| | - Jeong Kyu Bang
- Division of Magnetic Resonance, Korea Basic Science Institute (KBSI), Ochang, Cheongju 28119, Korea; (P.G.); (E.K.R.); (N.Y.P.); (Y.K.L.); (S.P.); (K.H.); (H.N.K.)
- Dandicure Inc., Ochang, Cheongju 28119, Korea
- Department of Bio-Analytical Science, University of Science & Technology, Daejeon 34113, Korea
- Correspondence: (Y.H.J.); (N.-K.S.); (J.K.B.)
| |
Collapse
|
3
|
Ramu D, Shan TW, Hirpara JL, Pervaiz S. Cellular senescence: Silent operator and therapeutic target in cancer. IUBMB Life 2021; 73:530-542. [PMID: 33675120 DOI: 10.1002/iub.2460] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 12/30/2022]
Abstract
The process of carcinogenesis and its progression involves an intricate interplay between a number of signaling networks, metabolic pathways and the microenvironment. These include an alteration in the cellular redox metabolism and deregulation of cell cycle checkpoints. Similar to the dichotomy of redox signaling in cancer cell fate and state determination, a diverging effect of an irreversible cell cycle arrest or senescence on carcinogenesis has been demonstrated. In this regard, while overwhelming oxidative stress has a damaging effect on tissue architecture and organ function and promotes death execution, a mild "pro-oxidant" environment is conducive for cell proliferation, growth and survival. Similarly, cellular senescence has been shown to elicit both a tumor suppressor and an oncogenic effect in a context-dependent manner. Notably, there appears to be a crosstalk between these two critical regulators of cell fate and state, particularly from the standpoint of the divergent effects on processes that promote or abate carcinogenesis. This review aims to provide an overview of these overarching themes and attempts to highlight critical intersection nodes, which are emerging as potential diagnostic and/or therapeutic targets for novel anticancer strategies.
Collapse
Affiliation(s)
- Deepika Ramu
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Teoh Wei Shan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jayshree L Hirpara
- Cancer Science Institute, National University of Singapore, Singapore, Singapore
| | - Shazib Pervaiz
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Medicine Healthy Longevity Program, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, National University Health System, Singapore, Singapore.,Integrative Science and Engineering Programme (ISEP), NUS Graduate School (NUSGS), National University of Singapore, Singapore, Singapore.,Faculté de Medicine, University of Paris, Paris, France
| |
Collapse
|
4
|
Castillo AF, Orlando UD, Maloberti PM, Prada JG, Dattilo MA, Solano AR, Bigi MM, Ríos Medrano MA, Torres MT, Indo S, Caroca G, Contreras HR, Marelli BE, Salinas FJ, Salvetti NR, Ortega HH, Lorenzano Menna P, Szajnman S, Gomez DE, Rodríguez JB, Podesta EJ. New inhibitor targeting Acyl-CoA synthetase 4 reduces breast and prostate tumor growth, therapeutic resistance and steroidogenesis. Cell Mol Life Sci 2021; 78:2893-2910. [PMID: 33068124 PMCID: PMC11072814 DOI: 10.1007/s00018-020-03679-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 09/15/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
Abstract
Acyl-CoA synthetase 4 (ACSL4) is an isoenzyme of the fatty acid ligase-coenzyme-A family taking part in arachidonic acid metabolism and steroidogenesis. ACSL4 is involved in the development of tumor aggressiveness in breast and prostate tumors through the regulation of various signal transduction pathways. Here, a bioinformatics analysis shows that the ACSL4 gene expression and proteomic signatures obtained using a cell model was also observed in tumor samples from breast and cancer patients. A well-validated ACSL4 inhibitor, however, has not been reported hindering the full exploration of this promising target and its therapeutic application on cancer and steroidogenesis inhibition. In this study, ACSL4 inhibitor PRGL493 was identified using a homology model for ACSL4 and docking based virtual screening. PRGL493 was then chemically characterized through nuclear magnetic resonance and mass spectroscopy. The inhibitory activity was demonstrated through the inhibition of arachidonic acid transformation into arachidonoyl-CoA using the recombinant enzyme and cellular models. The compound blocked cell proliferation and tumor growth in both breast and prostate cellular and animal models and sensitized tumor cells to chemotherapeutic and hormonal treatment. Moreover, PGRL493 inhibited de novo steroid synthesis in testis and adrenal cells, in a mouse model and in prostate tumor cells. This work provides proof of concept for the potential application of PGRL493 in clinical practice. Also, these findings may prove key to therapies aiming at the control of tumor growth and drug resistance in tumors which express ACSL4 and depend on steroid synthesis.
Collapse
Affiliation(s)
- Ana F Castillo
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET, Universidad de Buenos Aires, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ulises D Orlando
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET, Universidad de Buenos Aires, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Paula M Maloberti
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET, Universidad de Buenos Aires, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jesica G Prada
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET, Universidad de Buenos Aires, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Melina A Dattilo
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET, Universidad de Buenos Aires, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Angela R Solano
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET, Universidad de Buenos Aires, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María M Bigi
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET, Universidad de Buenos Aires, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mayra A Ríos Medrano
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET, Universidad de Buenos Aires, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María T Torres
- Departamento de Oncología Básico Clínico, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sebastián Indo
- Departamento de Oncología Básico Clínico, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Graciela Caroca
- Departamento de Oncología Básico Clínico, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Hector R Contreras
- Departamento de Oncología Básico Clínico, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Belkis E Marelli
- Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), CONICET, Universidad Nacional del Litoral, Esperanza, Santa Fe, Argentina
| | - Facundo J Salinas
- Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), CONICET, Universidad Nacional del Litoral, Esperanza, Santa Fe, Argentina
| | - Natalia R Salvetti
- Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), CONICET, Universidad Nacional del Litoral, Esperanza, Santa Fe, Argentina
| | - Hugo H Ortega
- Instituto de Ciencias Veterinarias del Litoral (ICiVet-Litoral), CONICET, Universidad Nacional del Litoral, Esperanza, Santa Fe, Argentina
| | - Pablo Lorenzano Menna
- Laboratorio de Oncología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Provincia de Buenos Aires, Argentina
| | - Sergio Szajnman
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Unidad de Microanálisis y Métodos Físicos Aplicados a Química Orgánica (UMYMFOR), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daniel E Gomez
- Laboratorio de Oncología Molecular, Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Provincia de Buenos Aires, Argentina
| | - Juan B Rodríguez
- Departamento de Química Orgánica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
- Unidad de Microanálisis y Métodos Físicos Aplicados a Química Orgánica (UMYMFOR), CONICET, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ernesto J Podesta
- Instituto de Investigaciones Biomédicas (INBIOMED), CONICET, Universidad de Buenos Aires, Paraguay 2155 (C1121ABG), Buenos Aires, Argentina.
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Armando RG, Gómez DLM, Gomez DE. New drugs are not enough‑drug repositioning in oncology: An update. Int J Oncol 2020; 56:651-684. [PMID: 32124955 PMCID: PMC7010222 DOI: 10.3892/ijo.2020.4966] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 12/16/2019] [Indexed: 11/24/2022] Open
Abstract
Drug repositioning refers to the concept of discovering novel clinical benefits of drugs that are already known for use treating other diseases. The advantages of this are that several important drug characteristics are already established (including efficacy, pharmacokinetics, pharmacodynamics and toxicity), making the process of research for a putative drug quicker and less costly. Drug repositioning in oncology has received extensive focus. The present review summarizes the most prominent examples of drug repositioning for the treatment of cancer, taking into consideration their primary use, proposed anticancer mechanisms and current development status.
Collapse
Affiliation(s)
- Romina Gabriela Armando
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Diego Luis Mengual Gómez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| | - Daniel Eduardo Gomez
- Laboratory of Molecular Oncology, Science and Technology Department, National University of Quilmes, Bernal B1876, Argentina
| |
Collapse
|
6
|
Structural Features of Nucleoprotein CST/Shelterin Complex Involved in the Telomere Maintenance and Its Association with Disease Mutations. Cells 2020; 9:cells9020359. [PMID: 32033110 PMCID: PMC7072152 DOI: 10.3390/cells9020359] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/29/2022] Open
Abstract
Telomere comprises the ends of eukaryotic linear chromosomes and is composed of G-rich (TTAGGG) tandem repeats which play an important role in maintaining genome stability, premature aging and onsets of many diseases. Majority of the telomere are replicated by conventional DNA replication, and only the last bit of the lagging strand is synthesized by telomerase (a reverse transcriptase). In addition to replication, telomere maintenance is principally carried out by two key complexes known as shelterin (TRF1, TRF2, TIN2, RAP1, POT1, and TPP1) and CST (CDC13/CTC1, STN1, and TEN1). Shelterin protects the telomere from DNA damage response (DDR) and regulates telomere length by telomerase; while, CST govern the extension of telomere by telomerase and C strand fill-in synthesis. We have investigated both structural and biochemical features of shelterin and CST complexes to get a clear understanding of their importance in the telomere maintenance. Further, we have analyzed ~115 clinically important mutations in both of the complexes. Association of such mutations with specific cellular fault unveils the importance of shelterin and CST complexes in the maintenance of genome stability. A possibility of targeting shelterin and CST by small molecule inhibitors is further investigated towards the therapeutic management of associated diseases. Overall, this review provides a possible direction to understand the mechanisms of telomere borne diseases, and their therapeutic intervention.
Collapse
|
7
|
Armando RG, Mengual Gomez DL, Maggio J, Sanmartin MC, Gomez DE. Telomeropathies: Etiology, diagnosis, treatment and follow-up. Ethical and legal considerations. Clin Genet 2019; 96:3-16. [PMID: 30820928 DOI: 10.1111/cge.13526] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 02/12/2019] [Accepted: 02/26/2019] [Indexed: 12/13/2022]
Abstract
Telomeropathies involve a wide variety of infrequent genetic diseases caused by mutations in the telomerase maintenance mechanism or the DNA damage response (DDR) system. They are considered a family of rare diseases that often share causes, molecular mechanisms and symptoms. Generally, these diseases are not diagnosed until the symptoms are advanced, diminishing the survival time of patients. Although several related syndromes may still be unrecognized this work describes those that are known, highlighting that because they are rare diseases, physicians should be trained in their early diagnosis. The etiology and diagnosis are discussed for each telomeropathy and the treatments when available, along with a new classification of this group of diseases. Ethical and legal issues related to this group of diseases are also considered.
Collapse
Affiliation(s)
- Romina G Armando
- Laboratory of Molecular Oncology, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Diego L Mengual Gomez
- Laboratory of Molecular Oncology, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Julián Maggio
- Laboratory of Molecular Oncology, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - María C Sanmartin
- Laboratory of Molecular Oncology, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Daniel E Gomez
- Laboratory of Molecular Oncology, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| |
Collapse
|
8
|
Wagner MS, Schultze E, Oliveira TL, de Leon PMM, Thurow HS, Campos VF, Oliveira I, de Souza D, Rodrigues OED, Collares T, Seixas FK. Revitalizing the AZT Through of the Selenium: An Approach in Human Triple Negative Breast Cancer Cell Line. Front Oncol 2018; 8:525. [PMID: 30524958 PMCID: PMC6262369 DOI: 10.3389/fonc.2018.00525] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 10/26/2018] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer represents about 15% of all cases of breast cancer, and still represents a therapeutic challenge. 3′-Azido-3′-deoxythymidine (AZT) is a nucleoside reverse transcriptase inhibitor with antitumor activity. Chalcogenides compounds, such as selenium, are very important intermediates applied in organic synthesis. Our objective was to investigate the effect and the underlying cell death mechanisms of AZT and its derivatives, in human breast cancer cell lines. The inhibitory effect of AZT and derivatives (1072, 1073, and 1079) was determined by MTT assay (0.1, 1, 10, 50, and 100 μM for concentrations and times 4, 24, 48, and 72 h) and Live/Dead in tumor cell lines MCF-7, MDA-MB 231 and also in non-tumor cell line CHO. Gene expression profiles related to apoptosis were investigated by qRT-PCR and induction of apoptosis was investigated by flow cytometry. MTT and Live/Dead assays showed that AZT derivatives decreased the rate of cell proliferation at concentrations of 50 and 100 μM in tumor cell lines MCF-7 and MDA-MB 231 while the commercial AZT presented a low antitumoral potential in all strains tested. In flow cytometry analysis we demonstrated that derivatives of AZT induced apoptosis, with an increase in both initial and late stages in both tumor cell lines evaluated, especially in MDA-MB 231. Our data show that the AZT derivative 1072 increased the expression of transcripts of the genes caspase 3 and 8 in MDA-MB 231 cell line when compared to control, suggesting that the extrinsic pathway of apoptosis was activated. In conclusion, derivatives of AZT, especially 1072, induce cytotoxicity in vitro in the triple negative breast cancer cell line through activation of the extrinsic pathway of apoptosis. These compounds containing selenium in its formulation are potential therapeutic agents for breast cancer.
Collapse
Affiliation(s)
- Mônica Silveira Wagner
- Programa de Pós-Graduação em Biotecnologia, Grupo de Pesquisa em Oncologia Celular e Molecular, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Eduarda Schultze
- Programa de Pós-Graduação em Biotecnologia, Grupo de Pesquisa em Oncologia Celular e Molecular, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Thais Larre Oliveira
- Programa de Pós-Graduação em Biotecnologia, Grupo de Pesquisa em Oncologia Celular e Molecular, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Priscila Marques Moura de Leon
- Programa de Pós-Graduação em Biotecnologia, Grupo de Pesquisa em Oncologia Celular e Molecular, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Helena Strelow Thurow
- Programa de Pós-Graduação em Biotecnologia, Grupo de Pesquisa em Oncologia Celular e Molecular, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Vinicius Farias Campos
- Programa de Pós-Graduação em Biotecnologia, Grupo de Pesquisa em Oncologia Celular e Molecular, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Isabel Oliveira
- Departamento de Fisiologia e Farmacologia, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Diego de Souza
- LabSelen-NanoBio - Universidade de Federal de Santa Maria, Santa Maria, Brazil
| | | | - Tiago Collares
- Programa de Pós-Graduação em Biotecnologia, Grupo de Pesquisa em Oncologia Celular e Molecular, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Fabiana Kömmling Seixas
- Programa de Pós-Graduação em Biotecnologia, Grupo de Pesquisa em Oncologia Celular e Molecular, Biotecnologia/Centro de Desenvolvimento Tecnológico, Universidade Federal de Pelotas, Pelotas, Brazil
| |
Collapse
|
9
|
Cardama GA, Gonzalez N, Maggio J, Menna PL, Gomez DE. Rho GTPases as therapeutic targets in cancer (Review). Int J Oncol 2017; 51:1025-1034. [PMID: 28848995 PMCID: PMC5592879 DOI: 10.3892/ijo.2017.4093] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/17/2017] [Indexed: 12/20/2022] Open
Abstract
Rho GTPases are key molecular switches controlling the transduction of external signals to cytoplasmic and nuclear effectors. In the last few years, the development of genetic and pharmacological tools has allowed a more precise definition of the specific roles of Rho GTPases in cancer. The aim of the present review is to describe the cellular functions regulated by these proteins with focus in deregulated signals present in malignant tumors. Finally, we describe the state of the art in search of different experimental therapeutic strategies with Rho GTPases as molecular targets.
Collapse
Affiliation(s)
- G A Cardama
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - N Gonzalez
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - J Maggio
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - P Lorenzano Menna
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| | - D E Gomez
- Laboratory of Molecular Oncology, Department of Science and Technology, Quilmes National University, Bernal B1876BXD, Buenos Aires, Argentina
| |
Collapse
|
10
|
Pitcovski J, Shahar E, Aizenshtein E, Gorodetsky R. Melanoma antigens and related immunological markers. Crit Rev Oncol Hematol 2017; 115:36-49. [DOI: 10.1016/j.critrevonc.2017.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 01/12/2023] Open
|
11
|
Wang H, Yu H, Sun Y, Zhao H, Guo Z, Yu B. Liriopesides B inhibited cell growth and decreased CA125 level in human ovarian cancer A2780 cells. Nat Prod Res 2017; 31:2198-2202. [PMID: 28449586 DOI: 10.1080/14786419.2017.1320788] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
To study the effect of liriopesides B on cell growth curve, cell doubling time, the activity of tumour marker CA125 and alkaline phosphatase (AKP) in human ovarian cancer A2780 cells. Both cell growth curve and doubling time were studied by MTT assay, CA125 level and AKP activity were determined by respective kits. Results showed that liriopesides B could shift down the A2780 cells growth curve in a dose-time-dependent manner and inhibit the proliferation in A2780 cells with the maximum inhibitory rate 94.462% at 120 h, the doubling time was prolonged too. CA125 level was decreased in a dose-dependent way as well as AKP activity. Liriopesides B exhibited potential anticancer activity against human ovarian cancer A2780 cells.
Collapse
Affiliation(s)
- Haiyan Wang
- a Department of Chemical Engineering and Food Science , Hubei University of Arts and Science , Hubei , China
| | - Haizhong Yu
- a Department of Chemical Engineering and Food Science , Hubei University of Arts and Science , Hubei , China
| | - Yonglin Sun
- a Department of Chemical Engineering and Food Science , Hubei University of Arts and Science , Hubei , China
| | - Huijun Zhao
- a Department of Chemical Engineering and Food Science , Hubei University of Arts and Science , Hubei , China
| | - Zhuang Guo
- a Department of Chemical Engineering and Food Science , Hubei University of Arts and Science , Hubei , China
| | - Bo Yu
- a Department of Chemical Engineering and Food Science , Hubei University of Arts and Science , Hubei , China
| |
Collapse
|
12
|
Contribution of Syncytins and Other Endogenous Retroviral Envelopes to Human Placenta Pathologies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 145:111-162. [DOI: 10.1016/bs.pmbts.2016.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|