1
|
Yadav AK, Maharjan Shrestha R, Yadav PN. Anticancer mechanism of coumarin-based derivatives. Eur J Med Chem 2024; 267:116179. [PMID: 38340509 DOI: 10.1016/j.ejmech.2024.116179] [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: 11/26/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
The structural motif of coumarins is related with various biological activities and pharmacological properties. Both natural coumarin extracted from various plants or a new coumarin derivative synthesized by modification of the basic structure of coumarin, in vitro experiments showed that coumarins are a promising class of anti-tumor agents with high selectivity. Cancer is a complex and multifaceted group of diseases characterized by the uncontrolled and abnormal growth of cells in the body. This review focuses on the anticancer mechanism of various coumarins synthesized and isolated in more than a decade. Isopentenyloxycoumarins inhibit angiogenesis by reducing CCl2 chemokine levels. Ferulin C is a potent colchicine-binding agent that destabilizes microtubules, exhibiting antiproliferative and anti-metastatic effects in breast cancer cells through PAK1 and PAK2-mediated signaling. Trimers of triphenylethylene-coumarin hybrids demonstrated significant proliferation inhibition in HeLa, A549, K562, and MCF-7 cell lines. Platinum(IV) complexes with 4-hydroxycoumarin have the potential for high genotoxicity against tumor cells, inducing apoptosis in SKOV-3 cells by up-regulating caspase 3 and caspase 9 expression. Derivatives of 3-benzyl coumarin seco-B-ring induce apoptosis, mediated through the PI3K/Akt/mTOR signaling pathway. Sesquiterpene coumarins inhibit the efflux pump of multidrug resistance-associated protein. Coumarin imidazolyl derivatives inhibit the aromatase enzyme, a major contributor to estrogen overproduction in estrogen-dependent breast cancer.
Collapse
Affiliation(s)
- Anand Kumar Yadav
- Central Department of Chemistry, Tribhuvan University, Kathmandu, Nepal
| | | | - Paras Nath Yadav
- Central Department of Chemistry, Tribhuvan University, Kathmandu, Nepal.
| |
Collapse
|
2
|
Bekkouche I, Shishonin AY, Vetcher AA. Recent Development in Biomedical Applications of Oligonucleotides with Triplex-Forming Ability. Polymers (Basel) 2023; 15:858. [PMID: 36850142 PMCID: PMC9964087 DOI: 10.3390/polym15040858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/12/2023] Open
Abstract
A DNA structure, known as triple-stranded DNA, is made up of three oligonucleotide chains that wind around one another to form a triple helix (TFO). Hoogsteen base pairing describes how triple-stranded DNA may be built at certain conditions by the attachment of the third strand to an RNA, PNA, or DNA, which might all be employed as oligonucleotide chains. In each of these situations, the oligonucleotides can be employed as an anchor, in conjunction with a specific bioactive chemical, or as a messenger that enables switching between transcription and replication through the triplex-forming zone. These data are also considered since various illnesses have been linked to the expansion of triplex-prone sequences. In light of metabolic acidosis and associated symptoms, some consideration is given to the impact of several low-molecular-weight compounds, including pH on triplex production in vivo. The review is focused on the development of biomedical oligonucleotides with triplexes.
Collapse
Affiliation(s)
- Incherah Bekkouche
- Nanotechnology Scientific and Educational Center, Institute of Biochemical Technology and Nanotechnology, Peoples’ Friendship University of Russia (RUDN), Miklukho-Maklaya Str. 6, Moscow 117198, Russia
| | - Alexander Y. Shishonin
- Complementary and Integrative Health Clinic of Dr. Shishonin, 5, Yasnogorskaya Str., Moscow 117588, Russia
| | - Alexandre A. Vetcher
- Nanotechnology Scientific and Educational Center, Institute of Biochemical Technology and Nanotechnology, Peoples’ Friendship University of Russia (RUDN), Miklukho-Maklaya Str. 6, Moscow 117198, Russia
- Complementary and Integrative Health Clinic of Dr. Shishonin, 5, Yasnogorskaya Str., Moscow 117588, Russia
| |
Collapse
|
3
|
Attina G, Mastrangelo S, Ruggiero A. Telomerase and Anticancer Treatment. BIOMEDICAL AND PHARMACOLOGY JOURNAL 2022; 15:1881-1888. [DOI: 10.13005/bpj/2526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Current chemotherapy uses compounds of organometallic nature that act with different mechanisms of action. Many pharmacological studies are directed toward the creation of compounds with more specific and selective activity toward tumor targets, including telomerase. The design and synthesis of such compounds with specific antitelomerase activity must consider the mechanism of action of the enzyme and its structure. The discovery of a close correlation between telomerase activation, cell immortalization and oncogenesis has suggested that telomerase inhibitors could be potent therapeutic agents, capable of selectively killing cancer cells. Inhibition of telomerase is expected to lead toward shortening of telomeres to a critical length, such that replicative senescence and cell death due to irreparable chromosomal damage can result. It has been observed that cancer cells generally have shorter telomeres than the normal replicative cell population, probably because the malignant cells have undergone more divisions. Therefore, the inhibition telomeres of cancer cells after a few cycles of cell division, without the normal cells suffering harmful consequences during therapy. Telomerase is certainly an interesting target on which to continue to study molecules that inhibit its function to obtain a specificity of therapeutic intervention and a reduction of the nonspecific cytotoxicity of chemotherapy.
Collapse
Affiliation(s)
- Giorgio Attina
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A.Gemelli IRCCS, Universita’ Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
4
|
A comprehensive review on acridone based derivatives as future anti-cancer agents and their structure activity relationships. Eur J Med Chem 2022; 239:114527. [PMID: 35717872 DOI: 10.1016/j.ejmech.2022.114527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 11/03/2022]
Abstract
The development of drug resistance and severe side-effects has reduced the clinical efficacy of the existing anti-cancer drugs available in the market. Thus, there is always a constant need to develop newer anti-cancer drugs with minimal adverse effects. Researchers all over the world have been focusing on various alternative strategies to discover novel, potent, and target specific molecules for cancer therapy. In this direction, several heterocyclic compounds are being explored but amongst them one promising heterocycle is acridone which has attracted the attention of medicinal chemists and gained huge biological importance as acridones are found to act on different therapeutically proven molecular targets, overcome ABC transporters mediated drug resistance and DNA intercalation in cancer cells. Some of these acridone derivatives have reached clinical studies as these heterocycles have shown huge potential in cancer therapeutics and imaging. Here, the authors have attempted to compile and make some recommendations of acridone based derivatives concerning their cancer biological targets and in vitro-cytotoxicity based on drug design and novelty to increase their therapeutic potential. This review also provides some important insights on the design, receptor targeting and future directions for the development of acridones as possible clinically effective anti-cancer agents.
Collapse
|
5
|
Recognition of ATT Triplex and DNA:RNA Hybrid Structures by Benzothiazole Ligands. Biomolecules 2022; 12:biom12030374. [PMID: 35327566 PMCID: PMC8945811 DOI: 10.3390/biom12030374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 11/17/2022] Open
Abstract
Interactions of an array of nucleic acid structures with a small series of benzothiazole ligands (bis-benzothiazolyl-pyridines—group 1, 2-thienyl/2-benzothienyl-substituted 6-(2-imidazolinyl)benzothiazoles—group 2, and three 2-aryl/heteroaryl-substituted 6-(2-imidazolinyl)benzothiazoles—group 3) were screened by competition dialysis. Due to the involvement of DNA:RNA hybrids and triplex helices in many essential functions in cells, this study’s main aim is to detect benzothiazole-based moieties with selective binding or spectroscopic response to these nucleic structures compared to regular (non-hybrid) DNA and RNA duplexes and single-stranded forms. Complexes of nucleic acids and benzothiazoles, selected by this method, were characterized by UV/Vis, fluorescence and circular dichroism (CD) spectroscopy, isothermal titration calorimetry, and molecular modeling. Two compounds (1 and 6) from groups 1 and 2 demonstrated the highest affinities against 13 nucleic acid structures, while another compound (5) from group 2, despite lower affinities, yielded higher selectivity among studied compounds. Compound 1 significantly inhibited RNase H. Compound 6 could differentiate between B- (binding of 6 dimers inside minor groove) and A-type (intercalation) helices by an induced CD signal, while both 5 and 6 selectively stabilized ATT triplex in regard to AT duplex. Compound 3 induced strong condensation-like changes in CD spectra of AT-rich DNA sequences.
Collapse
|
6
|
Morshedi K, Borran S, Ebrahimi MS, Masoud Khooy MJ, Seyedi ZS, Amiri A, Abbasi-Kolli M, Fallah M, Khan H, Sahebkar A, Mirzaei H. Therapeutic effect of curcumin in gastrointestinal cancers: A comprehensive review. Phytother Res 2021; 35:4834-4897. [PMID: 34173992 DOI: 10.1002/ptr.7119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/18/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Gastrointestinal (GI) cancers with a high global prevalence are a leading cause of morbidity and mortality. Accordingly, there is a great need to develop efficient therapeutic approaches. Curcumin, a naturally occurring agent, is a promising compound with documented safety and anticancer activities. Recent studies have demonstrated the activity of curcumin in the prevention and treatment of different cancers. According to systematic studies on curcumin use in various diseases, it can be particularly effective in GI cancers because of its high bioavailability in the gastrointestinal tract. Nevertheless, the clinical applications of curcumin are largely limited because of its low solubility and low chemical stability in water. These limitations may be addressed by the use of relevant analogues or novel delivery systems. Herein, we summarize the pharmacological effects of curcumin against GI cancers. Moreover, we highlight the application of curcumin's analogues and novel delivery systems in the treatment of GI cancers.
Collapse
Affiliation(s)
- Korosh Morshedi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sarina Borran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Zeynab Sadat Seyedi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Abbasi-Kolli
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Fallah
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
7
|
Awasthi A, Yadav P, Tiwari DK. A three-component, general and practical route for diastereoselective synthesis of aza-spirocyclic pyrazolones via a decarboxylative annulation process. NEW J CHEM 2021. [DOI: 10.1039/d0nj05915c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An efficient, general, and practical route for highly diastereoselective synthesis of aza-spirocyclic pyrazolones from easily available α-amino acids, aldehydes, and alkylidene pyrazolones by means of a decarboxylative annulation process is reported.
Collapse
Affiliation(s)
- Annapurna Awasthi
- Molecular Synthesis and Drug Discovery Unit
- Center of Biomedical Research (CBMR)
- SGPGIMS Campus
- Lucknow
- India
| | - Pushpendra Yadav
- Molecular Synthesis and Drug Discovery Unit
- Center of Biomedical Research (CBMR)
- SGPGIMS Campus
- Lucknow
- India
| | - Dharmendra Kumar Tiwari
- Molecular Synthesis and Drug Discovery Unit
- Center of Biomedical Research (CBMR)
- SGPGIMS Campus
- Lucknow
- India
| |
Collapse
|
8
|
Awasthi A, Yadav P, Kumar V, Tiwari DK. α‐Amino Acids Mediated C−C Double Bonds Cleavage in Diastereoselective Synthesis of Aza‐Spirocyclic Pyrazolones. Adv Synth Catal 2020. [DOI: 10.1002/adsc.202000884] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Annapurna Awasthi
- Molecular Synthesis and Drug Discovery Laboratory Center of Biomedical Research (CBMR) Sanjay Gandhi Post Graduate Institute of Medical Sciences Campus Raebareli Road Lucknow 226014 India
- Department of Chemistry Institute of Science Banaras Hindu University 221005 Varanasi Uttar Pradesh India
| | - Pushpendra Yadav
- Molecular Synthesis and Drug Discovery Laboratory Center of Biomedical Research (CBMR) Sanjay Gandhi Post Graduate Institute of Medical Sciences Campus Raebareli Road Lucknow 226014 India
- Department of Chemistry Institute of Science Banaras Hindu University 221005 Varanasi Uttar Pradesh India
| | - Virendra Kumar
- School of Basic Sciences Indian Institute of Technology Bhubaneswar Argul Khurdha 752050 Odisha India
| | - Dharmendra Kumar Tiwari
- Molecular Synthesis and Drug Discovery Laboratory Center of Biomedical Research (CBMR) Sanjay Gandhi Post Graduate Institute of Medical Sciences Campus Raebareli Road Lucknow 226014 India
| |
Collapse
|
9
|
Alexandrova E, Pecoraro G, Sellitto A, Melone V, Ferravante C, Rocco T, Guacci A, Giurato G, Nassa G, Rizzo F, Weisz A, Tarallo R. An Overview of Candidate Therapeutic Target Genes in Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12061470. [PMID: 32512900 PMCID: PMC7352306 DOI: 10.3390/cancers12061470] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/31/2020] [Accepted: 06/02/2020] [Indexed: 12/25/2022] Open
Abstract
Ovarian cancer (OC) shows the highest mortality rate among gynecological malignancies and, because of the absence of specific symptoms, it is frequently diagnosed at an advanced stage, mainly due to the lack of specific and early biomarkers, such as those based on cancer molecular signature identification. Indeed, although significant progress has been made toward improving the clinical outcome of other cancers, rates of mortality for OC are essentially unchanged since 1980, suggesting the need of new approaches to identify and characterize the molecular mechanisms underlying pathogenesis and progression of these malignancies. In addition, due to the low response rate and the high frequency of resistance to current treatments, emerging therapeutic strategies against OC focus on targeting single factors and pathways specifically involved in tumor growth and metastasis. To date, loss-of-function screenings are extensively applied to identify key drug targets in cancer, seeking for more effective, disease-tailored treatments to overcome lack of response or resistance to current therapies. We review here the information relative to essential genes and functional pathways recently discovered in OC, often strictly interconnected with each other and representing promising biomarkers and molecular targets to treat these malignancies.
Collapse
Affiliation(s)
- Elena Alexandrova
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
| | - Giovanni Pecoraro
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
| | - Assunta Sellitto
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
| | - Viola Melone
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
| | - Carlo Ferravante
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
- Genomix4Life, via S. Allende 43/L, 84081 Baronissi, Italy;
| | - Teresa Rocco
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
- Genomix4Life, via S. Allende 43/L, 84081 Baronissi, Italy;
| | - Anna Guacci
- Genomix4Life, via S. Allende 43/L, 84081 Baronissi, Italy;
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
- CRGS-Genome Research Center for Health, University of Salerno Campus of Medicine, 84081 Baronissi, Italy
- Correspondence: (A.W.); (R.T.); Tel.: +39-089-965043 (A.W.); +39-089-965067 (R.T.)
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitan”, University of Salerno, 84081 Baronissi, Italy; (E.A.); (G.P.); (A.S.); (V.M.); (C.F.); (T.R.); (G.G.); (G.N.); (F.R.)
- Correspondence: (A.W.); (R.T.); Tel.: +39-089-965043 (A.W.); +39-089-965067 (R.T.)
| |
Collapse
|
10
|
Zhang QD, Zhao BL, Li BY, Du DM. Squaramide-catalyzed asymmetric Mannich reactions between 3-fluorooxindoles and pyrazolinone ketimines. Org Biomol Chem 2019; 17:7182-7191. [PMID: 31322164 DOI: 10.1039/c9ob01350d] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
An enantioselective Mannich reaction between 3-fluorooxindoles and pyrazolinone ketimines has been developed for the construction of amino-pyrazolone-oxindoles containing stereogenic C-F units. Based on this new protocol that allows for the generation of two adjacent tetrasubstituted stereocenters, a variety of structurally diverse fluorinated amino-pyrazolone-oxindoles were obtained in good to excellent yields with excellent diastereoselectivities and enantioselectivities (up to 98% yield, >20 : 1 dr and >99% ee). What's more, good yield and high stereoselectivities were obtained in the gram-scale reaction.
Collapse
Affiliation(s)
- Qing-Da Zhang
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, 5 South Zhongguancun Street, Beijing 100081, P. R. China.
| | - Bo-Liang Zhao
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, 5 South Zhongguancun Street, Beijing 100081, P. R. China.
| | - Bing-Yu Li
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, 5 South Zhongguancun Street, Beijing 100081, P. R. China.
| | - Da-Ming Du
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, 5 South Zhongguancun Street, Beijing 100081, P. R. China.
| |
Collapse
|
11
|
DNA/RNA recognition controlled by the glycine linker and the guanidine moiety of phenanthridine peptides. Int J Biol Macromol 2019; 134:422-434. [PMID: 31082420 DOI: 10.1016/j.ijbiomac.2019.05.063] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 12/27/2022]
Abstract
The binding of four phenanthridine-guanidine peptides to DNA/RNA was evaluated via spectrophotometric/microcalorimetric methods and computations. The minor structural modifications-the type of the guanidine group (pyrrole guanidine (GCP) and arginine) and the linker length (presence or absence of glycine)-greatly affected the conformation of compounds and consequently the binding to double- (ds-) and single-stranded (ss-) polynucleotides. GCP peptide with shorter linker was able to distinguish between RNA (A-helix) and DNA (B-helix) by different circular dichroism response at 295 nm and thus can be used as a chiral probe. Opposed to the dominant stretched conformation of GCP peptide with shorter linker, the more flexible and longer linker of its analogue enabled the molecule to adopt the intramolecularly stacked form which resulted in weaker yet selective binding to DNA. Beside efficient organization of ss-polynucleotide structures, GCP peptide with shorter linker bound stronger to ss-DNA/RNA compared to arginine peptides which emphasize the importance of GCP unit.
Collapse
|
12
|
Zhu J, Liu W, Chen C, Zhang H, Yue D, Li C, Zhang L, Gao L, Huo Y, Liu C, Giaccone G, Zhang B, Wang C. TPP1 OB-fold domain protein suppresses cell proliferation and induces cell apoptosis by inhibiting telomerase recruitment to telomeres in human lung cancer cells. J Cancer Res Clin Oncol 2019; 145:1509-1519. [PMID: 31016380 DOI: 10.1007/s00432-019-02921-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/16/2019] [Indexed: 11/29/2022]
Abstract
PURPOSE Maintaining telomeres by recruiting telomerase-to-chromosome ends is essential for cancer cell survival. Inhibiting telomerase recruitment to telomeres represents a novel strategy for telomere-based lung cancer therapy. However, approaches for interrupting telomerase recruitment for cancer therapy still need to be explored. METHODS The telomere-binding protein TPP1 is responsible for recruiting telomerase to telomeres and synthesizing telomeres through the association between the oligosaccharide/oligonucleotide-binding (OB)-fold domain of TPP1 and telomerase reverse transcriptase. We overexpressed the TPP1 OB domain (TPP1-OB) by lentivirus infection in lung cancer cells. Telomere length was examined by Southern blot analysis of terminal restriction fragments. The effects of TPP1-OB on cell proliferation, the cell cycle, apoptosis, chemosensitivity, and tumor growth were evaluated in vitro and in vivo. RESULT TPP1-OB inhibited the recruitment of telomerase to telomeres and shortened telomere length by acting as a dominant-negative mutant of TPP1. TPP1-OB resulted in reduced cell proliferation, G1 cell cycle arrest, and increased cell apoptosis in lung cancer cells. Cell apoptosis occurred mainly through the caspase-3-dependent signaling pathway. TPP1-OB also suppressed anchorage-independent growth and tumor growth in vivo. Moreover, we demonstrated that TPP1-OB enhances the sensitivity of lung cancer cells to the chemotherapeutic drug paclitaxel. CONCLUSION Our results suggest that inhibiting TPP1-mediated telomerase recruitment by expressing the TPP1-OB domain is a potential novel strategy for telomere-targeted lung cancer therapy.
Collapse
Affiliation(s)
- Jinfang Zhu
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China
| | - Weiran Liu
- Department of Anesthesiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chen Chen
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China
| | - Hua Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China
| | - Dongsheng Yue
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China
| | - Chenguang Li
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China
| | - Lianmin Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China
| | - Liuwei Gao
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China
| | - Yansong Huo
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China
| | - Chang Liu
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China
| | - Giuseppe Giaccone
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China.,Georgetown University, Washington, DC, USA
| | - Bin Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China.
| | - Changli Wang
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Huan-Hu-Xi Road, Ti-Yuan-Bei, He Xi District, Tianjin, 300060, China.
| |
Collapse
|
13
|
Shukla K, Shah S, Rana NK, Singh VK. An efficient and highly diastereoselective synthesis of carbocyclic spiropyrazolones via one-pot sequential dual organo-silver catalyzed Michael-hydroalkylation reactions. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2018.11.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
14
|
Liu MY, Nemes A, Zhou QG. The Emerging Roles for Telomerase in the Central Nervous System. Front Mol Neurosci 2018; 11:160. [PMID: 29867352 PMCID: PMC5964194 DOI: 10.3389/fnmol.2018.00160] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Telomerase, a specialized ribonucleoprotein enzyme complex, maintains telomere length at the 3′ end of chromosomes, and functions importantly in stem cells, cancer and aging. Telomerase exists in neural stem cells (NSCs) and neural progenitor cells (NPCs), at a high level in the developing and adult brains of humans and rodents. Increasing studies have demonstrated that telomerase in NSCs/NPCs plays important roles in cell proliferation, neuronal differentiation, neuronal survival and neuritogenesis. In addition, recent works have shown that telomerase reverse transcriptase (TERT) can protect newborn neurons from apoptosis and excitotoxicity. However, to date, the link between telomerase and diseases in the central nervous system (CNS) is not well reviewed. Here, we analyze the evidence and summarize the important roles of telomerase in the CNS. Understanding the roles of telomerase in the nervous system is not only important to gain further insight into the process of the neural cell life cycle but would also provide novel therapeutic applications in CNS diseases such as neurodegenerative condition, mood disorders, aging and other ailments.
Collapse
Affiliation(s)
- Meng-Ying Liu
- Department of Clinical Pharmacology, Pharmacy College, Nanjing Medical University, Nanjing, China.,The Affiliated Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, China
| | - Ashley Nemes
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, Pharmacy College, Nanjing Medical University, Nanjing, China.,Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
15
|
Shariati M, Hajigholami S, Veisi Malekshahi Z, Entezari M, Bodaghabadi N, Sadeghizadeh M. Nanocurcumin-Mediated Down-Regulation of Telomerase Via Stimulating TGFβ1 Signaling Pathway in Hepatocellular Carcinoma Cells. IRANIAN BIOMEDICAL JOURNAL 2018; 22:171-9. [PMID: 28992682 PMCID: PMC5889502 DOI: 10.22034/ibj.22.3.171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background Curcumin, extracted from turmeric, represents enormous potential to serve as an anticancer agent. Telomerase is viewed as a prominent molecular target of curcumin, and Transforming growth factor-β1 (TGFβ1) has proven to be a major inhibitory signaling pathway for telomerase activity. In the current study, we aimed to explore suppressive effects of nanocurcumin on telomerase expression through TGFβ1 pathway in a hepatocellular carcinoma cell line (Huh7). Methods MTT assay was used to determine the effect of nonocurcumin on viability of Huh7 cells. RT-PCR was used to analyze the gene expression patterns. Results MTT assay revealed that nanocurcumin acts in a dose- and time-dependent manner to diminish the cell viability.
RT-PCR analysis indicated that nanocurcumin results in augmentation of TGFβ1 72 hours post treatment and leads to the reduction of telomerase expression 48 and 72 hours post exposure. Also, up-regulation of Smad3 and E2F1 and down-regulation of Smad7 confirmed the effect of nanocurcumin on intermediate components of TGFβ1 pathway. Furthermore, transfection of the proximal promoter of telomerase triggered a significant reduction in luciferase activity. Conclusion The data from the present study lead us to develop a deeper understanding of the mechanisms underlying nanocurcumin-mediated regulation of telomerase expression, thereby presenting a new perspective to the landscape of using nanocurcumin as a cancer-oriented therapeutic agent.
Collapse
Affiliation(s)
- Molood Shariati
- Department of Molecular Genetics, School of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-154, Tehran, Iran
| | - Samira Hajigholami
- Department of Molecular Genetics, School of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-154, Tehran, Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Entezari
- Department of Biology, Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | - Narges Bodaghabadi
- Department of Molecular Genetics, School of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-154, Tehran, Iran
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, School of Biological Sciences, Tarbiat Modares University, P.O. Box 14115-154, Tehran, Iran
| |
Collapse
|
16
|
Telomeres and Telomerase in Hematopoietic Dysfunction: Prognostic Implications and Pharmacological Interventions. Int J Mol Sci 2017; 18:ijms18112267. [PMID: 29143804 PMCID: PMC5713237 DOI: 10.3390/ijms18112267] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/15/2017] [Accepted: 10/24/2017] [Indexed: 12/31/2022] Open
Abstract
Leukocyte telomere length (TL) has been suggested as a marker of biological age in healthy individuals, but can also reflect inherited and acquired hematopoietic dysfunctions or indicate an increased turnover of the hematopoietic stem and progenitor cell compartment. In addition, TL is able to predict the response rate of tyrosine kinase inhibitor therapy in chronic myeloid leukemia (CML), indicates clinical outcomes in chronic lymphocytic leukemia (CLL), and can be used as screening tool for genetic sequencing of selected genes in patients with inherited bone marrow failure syndromes (BMFS). In tumor cells and clonal hematopoietic disorders, telomeres are continuously stabilized by reactivation of telomerase, which can selectively be targeted by telomerase-specific therapy. The use of the telomerase inhibitor Imetelstat in patients with essential thrombocythmia or myelofibrosis as well as the use of dendritic cell-based telomerase vaccination in AML patients with complete remissions are promising examples for anti-telomerase targeted strategies in hematologic malignancies. In contrast, the elevation in telomerase levels through treatment with androgens has become an exciting clinical intervention for patients with BMFS. Here, we review recent developments, which highlight the impact of telomeres and telomerase targeted therapies in hematologic dysfunctions.
Collapse
|
17
|
Anjong TF, Kim G, Jang HY, Yoon J, Kim J. Diiridium(iii) complexes: luminescent probes and sensors for G-quadruplex DNA and endoplasmic reticulum imaging. NEW J CHEM 2017. [DOI: 10.1039/c6nj02890j] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Two new dinuclear iridium (Ir) complexes bridged by a conjugated aromatic tppz ligand, (bhq)2Ir(tppz)Ir(bhq)2(1) and (ppy)2Ir(tppz)Ir(ppy)2(2) (bhq = benzo(h)quinolone, ppy = phenyl-pyridine, tppz = tetrapyrido[3,2-a:2′,3′-c:3′′,2′′-h:2′′′,3′′′-j]phenazine), were prepared.
Collapse
Affiliation(s)
| | - Gyoungmi Kim
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
| | - Ha Yoon Jang
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
| | - Juyoung Yoon
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
| | - Jinheung Kim
- Department of Chemistry and Nano Science
- Ewha Womans University
- Seoul 120-750
- Korea
| |
Collapse
|
18
|
RNA interference mediated downregulation of human telomerase reverse transcriptase (hTERT) in LN18 cells. Cytotechnology 2016; 68:2311-2321. [PMID: 27757712 DOI: 10.1007/s10616-016-0025-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 08/24/2016] [Indexed: 10/20/2022] Open
Abstract
Human telomerase reverse transcriptase (hTERT) gene is a biomarker for the targeted therapy in various cancers. Presence of increased telomerase activity is a common feature of all cancers including glioblastoma. Both RNA and catalytic subunits of hTERT are the target sites for blocking its activity. The current study focuses on the expression of hTERT in glioblastoma and its regulation using two different novel siRNAs (small interfering RNA). Our patient data demonstrated increased expression of hTERT, which could be correlated with carcinogenesis in glioma. In vitro studies in siRNA transfected LN18 cells confirmed significant cell death (p < 0.05) as evidenced by MTT and trypan blue exclusion assay. These results were further supported by flow cytometry data, which showed significant increase in early and late apoptosis. The hTERT mRNA expression was effectively downregulated by 45 and 39 % with siRNA1 and siRNA2, respectively. These results were further confirmed by immunoblotting analysis (p < 0.05). Our results suggest that both the siRNAs effectively down regulated the expression of hTERT at mRNA and protein levels, thereby decreasing cell viability and proliferation rate. Hence siRNA mediated downregulation of hTERT could be a potential therapeutic avenue in glioblastoma.
Collapse
|
19
|
Xue Y, Marvin ME, Ivanova IG, Lydall D, Louis EJ, Maringele L. Rif1 and Exo1 regulate the genomic instability following telomere losses. Aging Cell 2016; 15:553-62. [PMID: 27004475 PMCID: PMC4854909 DOI: 10.1111/acel.12466] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2016] [Indexed: 12/22/2022] Open
Abstract
Telomere attrition is linked to cancer, diabetes, cardiovascular disease and aging. This is because telomere losses trigger further genomic modifications, culminating with loss of cell function and malignant transformation. However, factors regulating the transition from cells with short telomeres, to cells with profoundly altered genomes, are little understood. Here, we use budding yeast engineered to lack telomerase and other forms of telomere maintenance, to screen for such factors. We show that initially, different DNA damage checkpoint proteins act together with Exo1 and Mre11 nucleases, to inhibit proliferation of cells undergoing telomere attrition. However, this situation changes when survivors lacking telomeres emerge. Intriguingly, checkpoint pathways become tolerant to loss of telomeres in survivors, yet still alert to new DNA damage. We show that Rif1 is responsible for the checkpoint tolerance and proliferation of these survivors, and that is also important for proliferation of cells with a broken chromosome. In contrast, Exo1 drives extensive genomic modifications in survivors. Thus, the conserved proteins Rif1 and Exo1 are critical for survival and evolution of cells with lost telomeres.
Collapse
Affiliation(s)
- Yuan Xue
- Newcastle University, Institute for Cell and Molecular Biosciences Institute for Cell and Molecular Biosciences (ICaMB) Newcastle upon Tyne UK
| | - Marcus E. Marvin
- Department of Genetics, Centre for Genetic Architecture of Complex Traits University of Leicester Leicester UK
| | - Iglika G. Ivanova
- Newcastle University, Institute for Cell and Molecular Biosciences Institute for Cell and Molecular Biosciences (ICaMB) Newcastle upon Tyne UK
| | - David Lydall
- Newcastle University, Institute for Cell and Molecular Biosciences Institute for Cell and Molecular Biosciences (ICaMB) Newcastle upon Tyne UK
| | - Edward J. Louis
- Department of Genetics, Centre for Genetic Architecture of Complex Traits University of Leicester Leicester UK
| | - Laura Maringele
- Newcastle University, Institute for Cell and Molecular Biosciences Institute for Cell and Molecular Biosciences (ICaMB) Newcastle upon Tyne UK
| |
Collapse
|
20
|
Li JH, Wen H, Liu L, Du DM. Diastereo- and Enantioselective Synthesis of Spiro-Pyrrolidine-Pyrazolones by Squaramide-Catalyzed Cascade Aza-Michael/Michael Reactions. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600316] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
21
|
Structural Insight into the interaction of Flavonoids with Human Telomeric Sequence. Sci Rep 2015; 5:17574. [PMID: 26627543 PMCID: PMC4667226 DOI: 10.1038/srep17574] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 11/03/2015] [Indexed: 12/13/2022] Open
Abstract
Flavonoids are a group of naturally available compounds that are an attractive source for drug discovery. Their potential to act as anti-tumourigenic and anti-proliferative agents has been reported previously but is not yet fully understood. Targeting human telomeric G-quadruplex DNA could be one of the mechanisms by which these flavonoids exert anticancer activity. We have performed detailed biophysical studies for the interaction of four representative flavonoids, Luteolin, Quercetin, Rutin and Genistein, with the human telomeric G-quadruplex sequence tetramolecular d-(T2AG3T) (Tel7). In addition, we used NMR spectroscopy to derive the first model for the complex formed between Quercetin and G-quadruplex sequence. The model showed that Quercetin stabilises the G-quadruplex structure and does not open the G-tetrad. It interacts with the telomeric sequence through π-stacking at two sites: between T1pT2 and between G6pT7. Based on our findings, we suggest that Quercetin could be a potent candidate for targeting the telomere and thus, act as a potent anti-cancer agent.
Collapse
|
22
|
Reddel RR. Telomere maintenance mechanisms in cancer: clinical implications. Curr Pharm Des 2015; 20:6361-74. [PMID: 24975603 PMCID: PMC4262939 DOI: 10.2174/1381612820666140630101047] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 06/26/2014] [Indexed: 01/20/2023]
Abstract
The presence of immortal cell populations with an up-regulated telomere maintenance mechanism (TMM) is an almost universal characteristic of cancers, whereas normal somatic cells are unable to prevent proliferation-associated telomere shortening and have a limited proliferative potential. TMMs and related aspects of telomere structure and function therefore appear to be ideal targets for the development of anticancer therapeutics. Such treatments would be targeted to a specific cancer-related molecular abnormality, and also be broad-spectrum in that they would be expected to be potentially applicable to most cancers. However, the telomere biology of normal and malignant human cells is a relatively young research field with large numbers of unanswered questions, so the optimal design of TMM-targeted therapeutic approaches remains unclear. This review outlines the opportunities and challenges presented by telomeres and TMMs for clinical management of cancer.
Collapse
Affiliation(s)
- Roger R Reddel
- Children's Medical Research Institute, 214 Hawkesbury Road, Westmead, New South Wales 2145, Australia.
| |
Collapse
|
23
|
Ghasemali S, Nejati-Koshki K, Tafsiri E, Rahmati-Yamchi M, Akbarzadeh A, Alizadeh E, Abbasi M, Barkhordari A, Tozihi M, Kordi S, Zarghami N. Inhibitory effects of β-cyclodextrin-helenalin complexes on H-TERT gene expression in the T47D breast cancer cell line - results of real time quantitative PCR. Asian Pac J Cancer Prev 2014; 14:6949-53. [PMID: 24377631 DOI: 10.7314/apjcp.2013.14.11.6949] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nowadays, the encapsulation of cytotoxic chemotherapeutic agents is attracting interest as a method for drug delivery. We hypothesized that the efficiency of helenalin might be maximized by encapsulation in β-cyclodextrin nanoparticles. Helenalin, with a hydrophobic structure obtained from flowers of Arnica chamissonis and Arnica Montana, has anti-cancer and anti-inflammatory activity but low water solubility and bioavailability. β-Cyclodextrin (β-CD) is a cyclic oligosaccharide comprising seven D-glucopyranoside units, linked through 1,4-glycosidic bonds. MATERIALS AND METHODS To test our hypothesis, we prepared β-cyclodextrin- helenalin complexes to determine their inhibitory effects on telomerase gene expression by real-time polymerase chain reaction (q-PCR) and cytotoxic effects by colorimetric cell viability (MTT) assay. RESULTS MTT assay showed that not only β-cyclodextrin has no cytotoxic effect on its own but also it demonstrated that β-cyclodextrin- helenalin complexes inhibited the growth of the T47D breast cancer cell line in a time and dose-dependent manner. Our q-PCR results showed that the expression of telomerase gene was effectively reduced as the concentration of β-cyclodextrin-helenalin complexes increased. CONCLUSIONS β-Cyclodextrin-helenalin complexes exerted cytotoxic effects on T47D cells through down-regulation of telomerase expression and by enhancing Helenalin uptake by cells. Therefore, β-cyclodextrin could be superior carrier for this kind of hydrophobic agent.
Collapse
Affiliation(s)
- Samaneh Ghasemali
- Drug Applied Research Center, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran E-mail : , ,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Le HT, Dean WL, Buscaglia R, Chaires JB, Trent JO. An investigation of G-quadruplex structural polymorphism in the human telomere using a combined approach of hydrodynamic bead modeling and molecular dynamics simulation. J Phys Chem B 2014; 118:5390-405. [PMID: 24779348 PMCID: PMC4032189 DOI: 10.1021/jp502213y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/21/2014] [Indexed: 01/12/2023]
Abstract
Guanine-rich oligonucleotides can adopt noncanonical tertiary structures known as G-quadruplexes, which can exist in different forms depending on experimental conditions. High-resolution structural methods, such as X-ray crystallography and NMR spectroscopy, have been of limited usefulness in resolving the inherent structural polymorphism associated with G-quadruplex formation. The lack of, or the ambiguous nature of, currently available high-resolution structural data, in turn, has severely hindered investigations into the nature of these structures and their interactions with small-molecule inhibitors. We have used molecular dynamics in conjunction with hydrodynamic bead modeling to study the structures of the human telomeric G-quadruplex-forming sequences at the atomic level. We demonstrated that molecular dynamics can reproduce experimental hydrodynamic measurements and thus can be a powerful tool in the structural study of existing G-quadruplex sequences or in the prediction of new G-quadruplex structures.
Collapse
Affiliation(s)
- Huy T. Le
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
| | - William L. Dean
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
- James
Graham Brown Cancer Center, University of
Louisville, 529 South
Jackson Street, Louisville, Kentucky 40202, United
States
| | - Robert Buscaglia
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
| | - Jonathan B. Chaires
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
- James
Graham Brown Cancer Center, University of
Louisville, 529 South
Jackson Street, Louisville, Kentucky 40202, United
States
- Department
of Medicine, School of Medicine, University
of Louisville, 550 South
Jackson Street, Louisville, Kentucky 40202, United
States
| | - John O. Trent
- Department
of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, HSC-A Building, Room 616, Louisville, Kentucky 40202, United States
- James
Graham Brown Cancer Center, University of
Louisville, 529 South
Jackson Street, Louisville, Kentucky 40202, United
States
- Department
of Medicine, School of Medicine, University
of Louisville, 550 South
Jackson Street, Louisville, Kentucky 40202, United
States
| |
Collapse
|
25
|
|
26
|
Synthesis, biological evaluation, and molecular docking studies of pyrazolyl-acylhydrazone derivatives as novel anticancer agents. Med Chem Res 2014. [DOI: 10.1007/s00044-014-0909-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
27
|
Shalaby T, Fiaschetti G, Nagasawa K, Shin-ya K, Baumgartner M, Grotzer M. G-quadruplexes as potential therapeutic targets for embryonal tumors. Molecules 2013; 18:12500-37. [PMID: 24152672 PMCID: PMC6269990 DOI: 10.3390/molecules181012500] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/18/2013] [Accepted: 09/25/2013] [Indexed: 12/27/2022] Open
Abstract
Embryonal tumors include a heterogeneous group of highly malignant neoplasms that primarily affect infants and children and are characterized by a high rate of mortality and treatment-related morbidity, hence improved therapies are clearly needed. G-quadruplexes are special secondary structures adopted in guanine (G)-rich DNA sequences that are often present in biologically important regions, e.g. at the end of telomeres and in the regulatory regions of oncogenes such as MYC. Owing to the significant roles that both telomeres and MYC play in cancer cell biology, G-quadruplexes have been viewed as emerging therapeutic targets in oncology and as tools for novel anticancer drug design. Several compounds that target these structures have shown promising anticancer activity in tumor xenograft models and some of them have entered Phase II clinical trials. In this review we examine approaches to DNA targeted cancer therapy, summarize the recent developments of G-quadruplex ligands as anticancer drugs and speculate on the future direction of such structures as a potential novel therapeutic strategy for embryonal tumors of the nervous system.
Collapse
Affiliation(s)
- Tarek Shalaby
- Division of Oncology, University Children's Hospital of Zurich, Zurich 8032, Switzerland.
| | | | | | | | | | | |
Collapse
|
28
|
You H, Lei M, Hu L. A green and efficient ammonium acetate-catalyzed one-pot synthesis of 4-[(2-hydroxynaphthalen-1-yl)(phenyl)methyl]-3-methyl-1H-pyrazol-5-ol derivatives. MONATSHEFTE FUR CHEMIE 2013. [DOI: 10.1007/s00706-013-1056-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
29
|
A Versatile Synthesis, PM3-Semiempirical, Antibacterial, and Antitumor Evaluation of Some Bioactive Pyrazoles. J Heterocycl Chem 2012. [DOI: 10.1002/jhet.806] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
30
|
Huang PR, Yeh YM, Pao CC, Chen CY, Wang TCV. N-(1-Pyrenyl) maleimide inhibits telomerase activity in a cell free system and induces apoptosis in Jurkat cells. Mol Biol Rep 2012; 39:8899-905. [PMID: 22707200 DOI: 10.1007/s11033-012-1757-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 06/07/2012] [Indexed: 02/04/2023]
Abstract
Telomerase activity is repressed in normal human somatic cells, but is activated in most cancers, suggesting that telomerase may be an important target for cancer therapy. Agents that interact selectively with telomerase are anticipated to exert specific action on cancer cells. In this study, we evaluated maleimide derivatives for their potency and selectivity of telomerase inhibition. Among the several N-substituted derivatives of maleimide tested, N-(1-Pyrenyl) maleimide was shown to exert the greatest inhibition of telomerase in a cell free system, with an IC50 value of 0.25 μM. Importantly, we demonstrated that N-(1-pyrenyl) maleimide induces apoptosis in Jurkat T cells and displays the greatest differential cytotoxicity against hematopoietic cancer cells. These results suggest that N-(1-pyrenyl) maleimide is an attractive maleimide to be tested and developed as anti-cancer drug.
Collapse
Affiliation(s)
- Pei-Rong Huang
- Department of Molecular and Cellular Biology, Chang Gung University, Kwei-San, Tao-Yuan, 333, Taiwan
| | | | | | | | | |
Collapse
|
31
|
Guittat L, Alberti P, Gomez D, De Cian A, Pennarun G, Lemarteleur T, Belmokhtar C, Paterski R, Morjani H, Trentesaux C, Mandine E, Boussin F, Mailliet P, Lacroix L, Riou JF, Mergny JL. Targeting human telomerase for cancer therapeutics. Cytotechnology 2011; 45:75-90. [PMID: 19003245 DOI: 10.1007/s10616-004-5127-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2004] [Accepted: 09/21/2004] [Indexed: 01/28/2023] Open
Abstract
The enzyme telomerase is involved in the replication of telomeres, specialized structures that cap and protect the ends of chromosomes. Its activity is required for maintenance of telomeres and for unlimited lifespan, a hallmark of cancer cells. Telomerase is overexpressed in the vast majority of human cancer cells and therefore represents an attractive target for therapy. Several approaches have been developed to inhibit this enzyme through the targeting of its RNA or catalytic components as well as its DNA substrate, the single-stranded 3'-telomeric overhang. Telomerase inhibitors are chemically diverse and include modified oligonucleotides as well as small diffusable molecules, both natural and synthetic. This review presents an update of recent investigations pertaining to these agents and discusses their biological properties in the context of the initial paradigm that the exposure of cancer cells to these agents should lead to progressive telomere shortening followed by a delayed growth arrest response.
Collapse
Affiliation(s)
- Lionel Guittat
- Laboratoire de Biophysique, Muséum National d'Histoire Naturelle USM503, INSERM U 565, CNRS UMR 5153, 43, rue Cuvier, 75231, Paris cedex 05, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Interaction of berberine, palmatine, coralyne, and sanguinarine to quadruplex DNA: A comparative spectroscopic and calorimetric study. Biochim Biophys Acta Gen Subj 2011; 1810:485-96. [DOI: 10.1016/j.bbagen.2011.01.011] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 01/21/2011] [Accepted: 01/24/2011] [Indexed: 12/11/2022]
|
33
|
Xu Y. Chemistry in human telomere biology: structure, function and targeting of telomere DNA/RNA. Chem Soc Rev 2011; 40:2719-40. [DOI: 10.1039/c0cs00134a] [Citation(s) in RCA: 249] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
34
|
Li H, Hua F, Zhao C, Liu G, Zhou Q. [Diagnotic value of the combined determination of telomerase activity in induced sputum, pleural effusion and fiberobronchoscopic biopsy samples in lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2010; 13:128-31. [PMID: 20673505 PMCID: PMC6000517 DOI: 10.3779/j.issn.1009-3419.2010.02.09] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 11/04/2009] [Indexed: 11/30/2022]
Abstract
背景与目的 已有的研究表明, 端粒酶的异常活化与肺癌的发生、发展及转归密切相关, 端粒酶是目前已知广谱的肿瘤分子标记物, 是诊断和治疗肺癌的重要分子标志物之一。本研究旨在探讨诱导痰、胸水和纤维支气管镜活检组织中端粒酶活性的联合检测对肺癌的诊断价值。 方法 对80例肺癌伴胸水患者和50例肺部良性病变伴胸水患者, 应用TRAP-PCR-ELISA方法分别检测其诱导痰、胸水和纤维支气管镜活检组织中端粒酶的活性。 结果 肺癌伴胸水患者诱导痰、胸水和纤维支气管镜活组织中端粒酶的活性均明显高于肺部良性病变伴胸水患者(P < 0.001), 不同病理类型的肺癌患者间三种标本的端粒酶活性差异无统计学意义(P > 0.05)。诱导痰、胸水和纤维支气管镜活组织中端粒酶活性的检测对肺癌的诊断敏感性分别为62.5%(50/80)、46.3%(37/80)和60.0%(48/80);特异性分别为72.0%(36/50)、66.0%(33/50)和70.0%(35/50);准确性分别为66.2%(86/130)、53.8%(70/130)和63.8%(83/130);三项联合检测的敏感性、特异性和准确性分别为85.0%(68/80)、78.0%(39/50)和82.3%(107/130), 其中敏感性与单独诱导痰、胸水和纤维支气管镜活组织的敏感性差异有统计学意义(P < 0.01)。 结论 诱导痰、胸水和纤维支气管镜活组织端粒酶活性的联合检测具有更高的敏感性, 可以提高肺癌的诊断率。
Collapse
Affiliation(s)
- Hongmei Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | | | | | | | | |
Collapse
|
35
|
Wesbuer S, Lanvers-Kaminsky C, Duran-Seuberth I, Bölling T, Schäfer KL, Braun Y, Willich N, Greve B. Association of telomerase activity with radio- and chemosensitivity of neuroblastomas. Radiat Oncol 2010; 5:66. [PMID: 20642823 PMCID: PMC2917444 DOI: 10.1186/1748-717x-5-66] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 07/19/2010] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Telomerase activity compensates shortening of telomeres during cell division and enables cancer cells to escape senescent processes. It is also supposed, that telomerase is associated with radio- and chemoresistance. In the here described study we systematically investigated the influence of telomerase activity (TA) and telomere length on the outcome of radio- and chemotherapy in neuroblastoma. METHODS We studied the effects on dominant negative (DN) mutant, wild type (WT) of the telomerase catalytic unit (hTERT) using neuroblastoma cell lines. The cells were irradiated with 60Co and treated with doxorubicin, etoposide, cisplatin and ifosfamide, respectively. Viability was determined by MTS/MTT-test and the GI50 was calculated. Telomere length was measured by southernblot analysis and TA by Trap-Assay. RESULTS Compared to the hTERT expressing cells the dominant negative cells showed increased radiosensitivity with decreased telomere length. Independent of telomere length, telomerase negative cells are significantly more sensitive to irradiation. The effect of TA knock-down or overexpression on chemosensitivity were dependent on TA, the anticancer drug, and the chemosensitivity of the maternal cell line. CONCLUSIONS Our results supported the concept of telomerase inhibition as an antiproliferative treatment approach in neuroblastomas. Telomerase inhibition increases the outcome of radiotherapy while in combination with chemotherapy the outcome depends on drug- and cell line and can be additive/synergistic or antagonistic. High telomerase activity is one distinct cancer stem cell feature and the here described cellular constructs in combination with stem cell markers like CD133, Aldehyddehydrogenase-1 (ALDH-1) or Side population (SP) may help to investigate the impact of telomerase activity on cancer stem cell survival under therapy.
Collapse
Affiliation(s)
- Simone Wesbuer
- Department of Radiotherapy -Radiooncology-, University Hospital Münster, Albert-Schweitzer-Strasse 33, D-48149 Münster
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Cai MY, Zhang B, He WP, Yang GF, Rao HL, Rao ZY, Wu QL, Guan XY, Kung HF, Zeng YX, Xie D. Decreased expression of PinX1 protein is correlated with tumor development and is a new independent poor prognostic factor in ovarian carcinoma. Cancer Sci 2010; 101:1543-9. [PMID: 20367640 PMCID: PMC11159430 DOI: 10.1111/j.1349-7006.2010.01560.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 02/23/2010] [Accepted: 02/25/2010] [Indexed: 01/11/2023] Open
Abstract
Human interacting protein X1 (PinX1) has been identified as a critical telomerase inhibitor and proposed to be a putative tumor suppressor gene. Loss of PinX1 has been found in a large variety of malignancies, but the expression status in epithelial ovarian tumors has not been investigated. In this study, immunohistochemistry for PinX1 protein was performed on a tissue microarray (TMA) of epithelial ovarian tumors (informatively containing 25 cystadenomas, 29 borderline tumors, and 157 invasive carcinomas) and 12 normal ovaries. Receiver-operator curve (ROC) analysis was used to determine cut-off scores for tumor positivity and to evaluate patients' survival status. The threshold for PinX1 positivity was determined to be above 60% (area under the curve = 0.856, P < 0.001) based on the area under the ROC. Positive expression of PinX1 was observed in 100% of normal ovarian tissues, in 84% of cystadenomas, in 75.9% borderline tumors, and 66.2% of ovarian carcinomas. Decreased expression of PinX1 was strongly related to patients with poor prognostic factors regarding presence of lymph node metastasis (P = 0.024), distant metastasis (P < 0.001), and late International Federation of Gynecology and Obstetrics (FIGO) stage (P < 0.001). In univariate survival analysis, a highly significant correlation between loss of PinX1 and shortened patient survival (mean, 48.2 months vs 99.2 months, P < 0.001) was displayed. Multivariate analysis demonstrated PinX1 expression (P = 0.027) was evaluated as an independent parameter. Our findings suggest that loss of PinX1 is an adverse independent molecular marker for epithelial ovarian carcinoma patients. PinX1 may be a novel target for telomerase-based anticancer therapy due to inhibiting telomerase activity.
Collapse
Affiliation(s)
- Mu-Yan Cai
- Sun Yat-Sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Masiero S, Trotta R, Pieraccini S, De Tito S, Perone R, Randazzo A, Spada GP. A non-empirical chromophoric interpretation of CD spectra of DNA G-quadruplex structures. Org Biomol Chem 2010; 8:2683-92. [PMID: 20440429 DOI: 10.1039/c003428b] [Citation(s) in RCA: 242] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
G-quadruplex DNA (G4-DNA) structures are four-stranded helical DNA (or RNA) structures, comprising stacks of G-tetrads, which are the outcome of planar association of four guanines in a cyclic Hoogsteen hydrogen-bonding arrangement. In the last decade the number of publications where CD spectroscopy has been used to study G4-DNAs, is extremely high. However, with very few exceptions, these investigations use an empirical interpretation of CD spectra. In this interpretation two basic types of CD spectra have been associated to a single specific difference in the features of the strand folding, i.e. the relative orientation of the strands, "parallel" (all strands have the same 5' to 3' orientation) or "antiparallel". Different examples taken from the literature where the empirical interpretation is not followed or is meaningless are presented and discussed. Furthermore, the case of quadruplexes formed by monomeric guanosine derivatives, where there is no strand connecting the adjacent quartets and the definition parallel/antiparallel strands cannot apply, will be discussed. The different spectral features observed for different G-quadruplexes is rationalised in terms of chromophores responsible for the electronic transitions. A simplified exciton coupling approach or more refined QM calculations allow to interpret the different CD features in terms of different stacking orientation (head-to-tail, head-to-head, tail-to-tail) between adjacent G-quartets irrespectively of the relative orientation of the stands (parallel/antiparallel).
Collapse
Affiliation(s)
- Stefano Masiero
- Alma Mater Studiorum-Università di Bologna, Dipartimento di Chimica Organica A. Mangini, via San Giacomo 11, I-40126, Bologna, Italy
| | | | | | | | | | | | | |
Collapse
|
38
|
Sohn JH, Yeh BI, Choi JW, Yoon J, Namkung J, Park KK, Kim HW. Repression of human telomerase reverse transcriptase using artificial zinc finger transcription factors. Mol Cancer Res 2010; 8:246-53. [PMID: 20145034 DOI: 10.1158/1541-7786.mcr-09-0141] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Telomerase activation is a key step in the development of human cancers. Expression of the catalytic subunit, human telomerase reverse transcriptase (hTERT), represents the limiting factor for telomerase activity. In this study, we have used artificial zinc finger protein (ZFP) transcription factors (TF) to repress the expression of hTERT in human cancer cell lines at the transcriptional level. We have constructed four-fingered ZFPs derived from the human genome which binds 12-bp recognition sequences within the promoter of the hTERT gene and fused them with a KRAB repressor domain to create a potent transcriptional repressor. Luciferase activity was decreased by >80% in all of the transcriptional repressors with luciferase reporter assay. When they were transfected into the telomerase-positive HEK293 cell line, a decrease of mRNA level and telomerase activity together with shortening of telomere length was observed. Actual growth of HEK293 cells was also inhibited by transfection of artificial ZFP-TFs. The repression was maintained for 100 days of culture. The repression of telomerase expression by artificial ZFP-TFs targeting the promoter region of the hTERT presents a new promising strategy for inhibiting the growth of human cancer cells.
Collapse
Affiliation(s)
- Joon Hyung Sohn
- Department of Biochemistry, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
39
|
Miri-Moghaddam E, Deezagi A, Soheili ZS. Downregulation of telomerase activity in human promyelocytic cell line using RNA interference. Ann Hematol 2009; 88:1169-76. [PMID: 19415276 DOI: 10.1007/s00277-009-0748-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2008] [Accepted: 04/20/2009] [Indexed: 11/24/2022]
Abstract
Telomerase is a ribonucleoprotein complex. It consists of two main components, human telomerase reverse transcriptase (hTERT) and human telomerase RNA. High telomerase activity is present in most malignant cells, but it is barely detectable in majority of somatic cells. The direct correlation between telomerase reactivation and carcinogens has made hTERT a key target for anticancer therapeutic studies. In this study, for the first time, we evaluated the ability of the new generation of short interfering RNA (siRNA) to regulate telomerase activity in the human promyelocytic leukemia cell line (HL-60). Transient transfection cell line by hTERT siRNAs resulted in statistically significant suppression of hTERT messenger RNAs which were detected by quantitative real-time polymerase chain reaction, while the expressed hTERT protein levels were measured by flow cytometry. The results of telomeric repeat amplification protocol showed that telomerase activity was significantly reduced upon transfection of the HL-60 cell line with hTERT siRNAs. The results of this study showed that telomerase activity and cell proliferation were efficiently inhibited in the hTERT siRNA-treated leukemic cell line.
Collapse
Affiliation(s)
- E Miri-Moghaddam
- National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | | | | |
Collapse
|
40
|
DNA adducts of antitumor cisplatin preclude telomeric sequences from forming G quadruplexes. J Biol Inorg Chem 2009; 14:959-68. [PMID: 19390878 DOI: 10.1007/s00775-009-0508-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2009] [Accepted: 04/16/2009] [Indexed: 10/20/2022]
Abstract
We studied the effect of antitumor cisplatin and inefficient transplatin on the structure and stability of G quadruplexes formed by the model human telomere sequence 5'-GGG(TTAGGG)(3)-3' using circular dichroism, UV-monitored thermal denaturation, and gel electrophoresis. In addition, to investigate whether there is a connection between the ability of cisplatin or transplatin to affect telomerase activity and stability of G quadruplexes, we also used a modified telomere repeat amplification protocol assay that uses an oligonucleotide substrate for telomerase elongation susceptible to forming a G quadruplex. The results indicate that cisplatin is more efficient than transplatin in disturbing the quadruplex structure, thereby precluding telomeric sequences from forming quadruplexes. On the other hand, the results of this work also demonstrate that in absence of free platinum complex, DNA adducts of antitumor cisplatin inhibit telomerase catalysis, so the mechanism underlying this inhibition does not involve formation of the G quadruplexes which are not elongated by telomerase.
Collapse
|
41
|
Liu H, Gauld JW. Protonation of guanine quartets and quartet stacks: insights from DFT studies. Phys Chem Chem Phys 2009; 11:278-87. [DOI: 10.1039/b811717a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
42
|
Zhang B, Bai YX, Ma HH, Feng F, Jin R, Wang ZL, Lin J, Sun SP, Yang P, Wang XX, Huang PT, Huang CF, Peng Y, Chen YC, Kung HF, Huang JJ. Silencing PinX1 Compromises Telomere Length Maintenance As Well As Tumorigenicity in Telomerase-Positive Human Cancer Cells. Cancer Res 2008; 69:75-83. [DOI: 10.1158/0008-5472.can-08-1393] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
43
|
Melatonin down-regulates hTERT expression induced by either natural estrogens (17β-estradiol) or metalloestrogens (cadmium) in MCF-7 human breast cancer cells. Cancer Lett 2008; 268:272-7. [DOI: 10.1016/j.canlet.2008.04.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2008] [Revised: 04/01/2008] [Accepted: 04/02/2008] [Indexed: 11/24/2022]
|
44
|
Qu Y, Zhang L, Mao M, Zhao F, Huang X, Yang C, Xiong Y, Mu D. Effects of DNAzymes targeting Aurora kinase A on the growth of human prostate cancer. Cancer Gene Ther 2008; 15:517-25. [PMID: 18404163 DOI: 10.1038/cgt.2008.22] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aurora kinase A has been demonstrated to be involved in the malignant progression of many types of cancer including prostate cancer, we therefore hypothesized that Aurora kinase A might work as a valuable target for prostate cancer treatment. To test this hypothesis, we used DNAzyme technology to inhibit Aurora kinase A expression and evaluated the effects of DNAzymes as therapeutic agents to treat prostate cancer. In an in vitro cleavage assay, we found that a DNAzyme (DZ2) targeting Aurora kinase A could effectively cleave Aurora kinase A mRNA. When transfected into the prostate cancer cell line PC3, DZ2 was found to strongly inhibit the expression of Aurora kinase A examined by western blot analysis, and thus suppressed cell growth, arrested the progression of cell cycle, induced cell apoptosis and attenuated cell migration, as measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium assay, flow cytometry and Boyden chamber assay. Through in vivo study, we also found that DZ2 could significantly inhibit the growth of human prostate cancer xenografts in nude mice. In conclusion, DZ2 could effectively attenuate malignant progression of prostate cancer both in vitro and in vivo, suggesting that DNAzyme targeting Aurora kinase A may be used as a valuable therapy to treat prostate cancer.
Collapse
Affiliation(s)
- Y Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Paramasivan S, Rujan I, Bolton PH. Circular dichroism of quadruplex DNAs: applications to structure, cation effects and ligand binding. Methods 2008; 43:324-31. [PMID: 17967702 DOI: 10.1016/j.ymeth.2007.02.009] [Citation(s) in RCA: 427] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Accepted: 02/14/2007] [Indexed: 10/22/2022] Open
Abstract
Circular dichroism, CD, spectra can be used to gain information about quadruplex structures of DNAs as well as the effects of sequence, cations, chemical modification and ligand binding on quadruplex structure. There is not yet a validated approach to calculate a CD spectrum from a quadruplex structure nor is their one to go from a CD spectrum to a structure. However, it is possible to empirically correlate CD spectra features with quadruplex structural type in many cases. In this article four case studies are presented to indicate the strengths and limitations of CD in investigations of the properties of quadruplex structures formed by telomere repeat sequences. The case studies include determination of the quadruplex structural type present as a function of potassium concentration, the effect of sequence on the equilibrium between quadruplex structural types as a function of potassium concentration, the effect of ligand binding on quadruplex structure and the effect of 5' phosphorylation on quadruplex structural type.
Collapse
|
46
|
Bu X, Jia F, Wang W, Guo X, Wu M, Wei L. Coupled down-regulation of mTOR and telomerase activity during fluorouracil-induced apoptosis of hepatocarcinoma cells. BMC Cancer 2007; 7:208. [PMID: 17996122 PMCID: PMC2186345 DOI: 10.1186/1471-2407-7-208] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Accepted: 11/12/2007] [Indexed: 11/16/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most invasive and frequently diagnosed malignancy and the second leading cause of cancer death in many regions of Asia. The PI3K/Akt/mTOR signal pathway is involved in multiple cellular functions including proliferation, differentiation, tumorigenesis, and apoptosis. Up-regulation of telomerase activity is thought to be a critical step leading to cell transformation. Methods This study investigated changes in mTOR pathway and telomerase activity in hepatocarcinoma cell line SMMC-7721 treated with chemotherapeutic agent 5-fluorouracil (5-Fu). We detected apoptosis of hepatocarcinoma cells by TUNEL assay. Telomerase activity, hTERT transcription level and p- p70 S6k was demonstrated by the telomeric repeat amplification protocol and silver staining assay, Dual-Luciferase Reporter Assay and Western blot analysis respectively. Results Treating SMMC-7721 cells with 5-Fu leads to apoptosis of the cells, and reduction in telomerase activity, as well as a dramatic reduction in the activated form of p70 S6 kinase, a mTOR substrate. The 5-Fu treatment nearly abolishes transcription of hTERT (the major component of telomerase) mRNA. Treating SMMC-7721 cells with Rapamycin, a specific mTOR inhibitor, significantly reduce hTERT protein level but did not affect hTERT transcription. 5-Fu and rapamycin were synergistic in regards to down-regulation of telomerase activity in hepatocarcinoma cells. Conclusion These results suggest that chemotherapeutic agent 5-Fu may down-regulate telomerase activity at both transcriptional level and PI3K/Akt/mTOR pathway-dependent post-transcriptional level to facilitate hepatocellular carcinoma cell apoptosis.
Collapse
Affiliation(s)
- Xinxin Bu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Hospital, Second Military Medical Universisty, 225 Changhai Road, Shanghai 200438, China.
| | | | | | | | | | | |
Collapse
|
47
|
Shariftabrizi A, Eller MS. Telomere homolog oligonucleotides and the skin: current status and future perspectives. Exp Dermatol 2007; 16:627-33. [PMID: 17620088 DOI: 10.1111/j.1600-0625.2007.00580.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Ahmad Shariftabrizi
- Department of Dermatology, Boston University School of Medicine, Boston, MA 02118, USA
| | | |
Collapse
|
48
|
Roy S, Tanious FA, Wilson WD, Ly DH, Armitage BA. High-affinity homologous peptide nucleic acid probes for targeting a quadruplex-forming sequence from a MYC promoter element. Biochemistry 2007; 46:10433-43. [PMID: 17718513 DOI: 10.1021/bi700854r] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Guanine-rich DNA and RNA sequences are known to fold into secondary structures known as G-quadruplexes. Recent biochemical evidence along with the discovery of an increasing number of sequences in functionally important regions of the genome capable of forming G-quadruplexes strongly indicates important biological roles for these structures. Thus, molecular probes that can selectively target quadruplex-forming sequences (QFSs) are envisioned as tools to delineate biological functions of quadruplexes as well as potential therapeutic agents. Guanine-rich peptide nucleic acids have been previously shown to hybridize to homologous DNA or RNA sequences forming PNA-DNA (or RNA) quadruplexes. For this paper we studied the hybridization of an eight-mer G-rich PNA to a quadruplex-forming sequence derived from the promoter region of the MYC proto-oncogene. UV melting analysis, fluorescence assays, and surface plasmon resonance experiments reveal that this PNA binds to the MYC QFS in a 2:1 stoichiometry and with an average binding constant Ka = (2.0 +/- 0.2) x 10(8) M(-1) or Kd = 5.0 nM. In addition, experiments carried out with short DNA targets revealed a dependence of the affinity on the sequence of bases in the loop region of the DNA. A structural model for the hybrid quadruplex is proposed, and implications for gene targeting by G-rich PNAs are discussed.
Collapse
Affiliation(s)
- Subhadeep Roy
- Department of Chemistry, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, Pennsylvania 15213-3890, USA
| | | | | | | | | |
Collapse
|
49
|
Li Y, Li H, Yao G, Li W, Wang F, Jiang Z, Li M. Inhibition of telomerase RNA (hTR) in cervical cancer by adenovirus-delivered siRNA. Cancer Gene Ther 2007; 14:748-55. [PMID: 17479103 DOI: 10.1038/sj.cgt.7701056] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Small interfering RNA (siRNA) has become a powerful tool for selectively silencing gene expression in cultured mammalian cells. In this study, a 67-bp oligonucleotide encoding human telomerase RNA (hTR) was introduced into pSIREN, a shuttle vector for construction of recombinant adenovirus. Then the U6-RNA promoter and siRNA-encoding insert were cut out from the pSIREN and subcloned into pAdeno-X to construct the plasmid pAd-hTR. After the pAd-hTR was transfected into a mammalian cell line HEK-293, adenovirus carrying the hTR-targeting siRNA (Ad-hTR-siRNA) was obtained. We performed a series of experiments to demonstrate silencing of the telomerase mediated by Ad-hTR-siRNA in HeLa cells. Compared with control virus (Ad-NT-siRNA), Ad-hTR-siRNA significantly reduced both hTR mRNA level (by 70.21%) and telomerase activity (by 58.87%) in HeLa cells. Moreover, it induced apoptosis in HeLa cells. Treatment of subcutaneous tumor xenografted with Ad-hTR-siRNA could slow down tumor growth, at least partially due to the induction of apoptosis (P<0.05) in vivo. Taken together, our results demonstrated efficient and specific knockdown of telomerase in HeLa cell line by the hTR siRNA, and indicated the prospect of applying this siRNA expressing recombinant adenovirus system in cancer gene therapy.
Collapse
Affiliation(s)
- Y Li
- Department of Microbiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | | | | | | | | | | | | |
Collapse
|
50
|
Zhong C, Wang J, Wu N, Wu G, Zavalij PY, Shi X. Anion bridged nanosheet from self-assembled G-quadruplexes. Chem Commun (Camb) 2007:3148-50. [PMID: 17653370 DOI: 10.1039/b704756h] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A novel, non-covalent polymeric nanosheet has been produced through small molecule self-assembly; the structure has been characterized by solution NMR, solid-state NMR, powder XRD and AFM.
Collapse
Affiliation(s)
- Cheng Zhong
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506, USA
| | | | | | | | | | | |
Collapse
|