1
|
Pant P. Design and Characterization of Neutral Linker-Based Bis-Intercalator via Computer Simulations: Balancing DNA Binding and Cellular Uptake. Chem Biodivers 2024; 21:e202400768. [PMID: 38980964 DOI: 10.1002/cbdv.202400768] [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: 04/09/2024] [Revised: 06/23/2024] [Accepted: 07/09/2024] [Indexed: 07/11/2024]
Abstract
Bis-intercalators refer to a class of chemical compounds known for their unique ability to simultaneously intercalate, or insert, into DNA at two distinct sites. These molecules typically feature two intercalating moieties connected by a linker, allowing them to engage with DNA base pairs at multiple locations. The bis-intercalation phenomenon plays a significant role in altering the DNA structure, affecting its stability, and potentially influencing various cellular processes. These compounds have gained considerable attention in medicinal chemistry and biochemistry due to their potential applications in cancer therapy, where they may interfere with DNA replication and transcription, leading to anticancer effects. Traditionally, these molecules often possess a high positive charge to enhance their affinity for the negatively charged DNA. However, due to a high positive charge, their cellular uptake is compromised, along with their enhanced potential off-target effects. In this study, we utilized bis-intercalator TOTO and replaced the charged linker segment (propane-1,3-diammonium) with a neutral peroxodisulphuric acid linker. Using molecular modeling and computer simulations (500 ns, 3 replicas), we investigated the potential of the designed molecule as a bis-intercalator and compared the properties with the control bis-intercalator bound to DNA. We observed that the designed bis-intercalator exhibited improved DNA binding (as assessed through MM-PBSA and Delphi methods) and membrane translocation permeability. With an overall reduced charge, significantly less off-target binding of the designed molecule is also anticipated. Consequently, bis-intercalators based on peroxodisulphuric linkers can potentially target DNA effectively, and their role in the future design of bis-intercalators is foreseen.
Collapse
Affiliation(s)
- Pradeep Pant
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, UP, India
| |
Collapse
|
2
|
Mashita R, Sakae H, Nishiyama Y, Nagatani H. Spectroelectrochemical Analysis of Ion Transfer Mechanisms of Mitoxantrone at Liquid|Liquid Interfaces: Effects of Zwitterionic Dendrimer and Phospholipid Layer. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:2111-2119. [PMID: 38171364 DOI: 10.1021/acs.langmuir.3c02980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The ionic partition property and transfer mechanism of the anthraquinone antitumor agent mitoxantrone (MTX) were studied in detail at the water|1,2-dichloroethane (DCE) interface by means of surface-sensitive spectroelectrochemical techniques. The interfacial mechanism of the cationic MTX species was composed of potential-driven ion transfer and adsorption processes. The ion association between MTX and zwitterionic polyamidoamine (PAMAM) dendrimers with peripheral carboxy groups was also investigated in terms of the effects of pH and dendritic generation. The monovalent HMTX+ interacted effectively with the negatively charged dendrimers at neutral pH, while the divalent H2MTX2+ exhibited a weak association under acidic conditions. The higher stability of the dendrimer-MTX associates in the interfacial region was found for higher dendritic generations: G3.5 ≥ G2.5 > G1.5. The interfacial behavior of MTX and its dendrimer associates was further analyzed at the phospholipid-modified interface as a model biomembrane surface. The adsorption process of HMTX+ occurred mainly on the hydrophilic side of the phospholipid layer. The spectroelectrochemical results indicated that the dendrimers penetrate into the phospholipid layer and alter the transfer mechanism of HMTX+ across the interface.
Collapse
Affiliation(s)
- Ryuto Mashita
- Division of Material Chemistry, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Hiroki Sakae
- Division of Material Chemistry, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
- Faculty of Chemistry, Institute of Science and Engineering, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Yoshio Nishiyama
- Division of Material Chemistry, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
- Faculty of Chemistry, Institute of Science and Engineering, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| | - Hirohisa Nagatani
- Division of Material Chemistry, Graduate School of Natural Science and Technology, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
- Faculty of Chemistry, Institute of Science and Engineering, Kanazawa University, Kakuma, Kanazawa 920-1192, Japan
| |
Collapse
|
3
|
Amiran MR, Taghdir M, Abasi Joozdani F. Investigation of the inhibitory behavior of XFE and mitoxantrone molecules in interaction with AKT1 protein: a molecular dynamics simulation study. J Mol Model 2023; 29:153. [PMID: 37086344 DOI: 10.1007/s00894-023-05520-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/17/2023] [Indexed: 04/23/2023]
Abstract
The PI3K/Akt/mTOR pathway is one of the important pathways in many cancers. Akt is a serine-threonine kinase protein identified as a drug target for cancer treatment. Therefore, anticancer drugs are essential therapeutic targets for this pathway. In the current study, the inhibitory effect of two anticancer molecules, XFE and mitoxantrone, on AKT1 protein that can impact the activity of the AKT1 protein was investigated by using molecular docking and molecular dynamics (MD) simulations. The molecular docking results presented a relatively higher binding affinity of the mitoxantrone molecule in interaction with AKT1 than the XFE molecule. These results were validated by the MM/PBSA technique that was performed on obtained trajectories of 25 ns MD simulations. The mitoxantrone molecule has an intense binding energy of - 880.536 kcal/mol with AKT1 protein, while the XFE molecule shows a binding energy value of - 83.569 kcal/mol. Our findings from molecular dynamics simulations indicated that both molecules have favorite interactions with AKT1 protein. Other analyses, such as RMSF and hydrogen binding on trajectories obtained from MD simulations, indicated that the mitoxantrone molecule could be a relatively potent inhibitor for AKT1. Based on the results of this study and the structure of mitoxantrone, it is expected to be a good candidate for cancer treatment as a (PI3K)/Akt/mTOR inhibitor.
Collapse
Affiliation(s)
- Mohammad Reza Amiran
- Department of Biophysics, Faculty of Biological Science, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Majid Taghdir
- Department of Biophysics, Faculty of Biological Science, Tarbiat Modares University, Tehran, 14115-111, Iran.
| | - Farzane Abasi Joozdani
- Department of Biophysics, Faculty of Biological Science, Tarbiat Modares University, Tehran, 14115-111, Iran
| |
Collapse
|
4
|
Marzęda P, Wróblewska-Łuczka P, Drozd M, Florek-Łuszczki M, Załuska-Ogryzek K, Łuszczki JJ. Cannabidiol Interacts Antagonistically with Cisplatin and Additively with Mitoxantrone in Various Melanoma Cell Lines-An Isobolographic Analysis. Int J Mol Sci 2022; 23:ijms23126752. [PMID: 35743195 PMCID: PMC9224300 DOI: 10.3390/ijms23126752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/11/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
The medical application of cannabidiol (CBD) has been gathering increasing attention in recent years. This non-psychotropic cannabis-derived compound possesses antiepileptic, antipsychotic, anti-inflammatory and anxiolytic properties. Recent studies report that it also exerts antineoplastic effects in multiple types of cancers, including melanoma. In this in vitro study we tried to reveal the anticancer properties of CBD in malignant melanoma cell lines (SK-MEL 28, A375, FM55P and FM55M2) administered alone, as well as in combination with mitoxantrone (MTX) or cisplatin (CDDP). The effects of CBD on the viability of melanoma cells were measured by the MTT assay; cytotoxicity was determined in the LDH test and proliferation in the BrdU test. Moreover, the safety of CBD was tested in human keratinocytes (HaCaT) in LDH and MTT tests. Results indicate that CBD reduces the viability and proliferation of melanoma-malignant cells and exerts additive interactions with MTX. Unfortunately, CBD produced antagonistic interaction when combined with CDDP. CBD does not cause significant cytotoxicity in HaCaT cell line. In conclusion, CBD may be considered as a part of melanoma multi-drug therapy when combined with MTX. A special attention should be paid to the combination of CBD with CDDP due to the antagonistic interaction observed in the studied malignant melanoma cell lines.
Collapse
Affiliation(s)
- Paweł Marzęda
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
| | - Paula Wróblewska-Łuczka
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
| | - Małgorzata Drozd
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
| | | | - Katarzyna Załuska-Ogryzek
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
| | - Jarogniew J. Łuszczki
- Department of Pathophysiology, Medical University of Lublin, 20-090 Lublin, Poland; (P.M.); (P.W.-Ł.); (M.D.); (K.Z.-O.)
- Correspondence: ; Tel.: +48-81-448-6500; Fax: +48-81-448-6501
| |
Collapse
|
5
|
Zhang Y, Liu Q, Wei W, Zhang G, Yan S, Dai R, Sun Y, Su D, Lv S, Xia Y, Li J, Li C. Bortezomib potentiates antitumor activity of mitoxantrone through dampening Wnt/β-catenin signal pathway in prostate cancer cells. BMC Cancer 2021; 21:1101. [PMID: 34645397 PMCID: PMC8515742 DOI: 10.1186/s12885-021-08841-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 10/05/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Bortezomib (BZM), alone or in combination with other chemotherapies, has displayed strong anticancer effects in several cancers. The efficacy of the combination of BZM and mitoxantrone (MTX) in treating prostate cancer remains unknown. METHODS Anticancer effects of combination of BZM and MTX were determined by apoptosis and proliferation assay in vivo and in vitro. Expression of β-Catenin and its target genes were characterized by western blot and Real-time PCR. RESULTS BZM significantly enhanced MTX-induced antiproliferation in vivo and in vitro. Mice administered a combination of BZM and MTX displayed attenuated tumor growth and prolonged survival. BZM significantly attenuated MTX-induced apoptosis. Moreover, the combination of BZM and MTX contributed to inhibition of the Wnt/β-Catenin signaling pathway compared to monotherapy. CONCLUSIONS This study demonstrates that BZM enhances MTX-induced anti-tumor effects by inhibiting the Wnt/β-Catenin signaling pathway in prostate cancer cells.
Collapse
Affiliation(s)
- Ying Zhang
- Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Qiuzi Liu
- Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Wei Wei
- Center for Experimental Medicine, School of Public Health, Jining Medical University, Jining, 272067, China
| | - Guoan Zhang
- Institute of Cancer Pathology Research, Jining Medical University, Jining, 272067, China
| | - Siyuan Yan
- Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Rongrong Dai
- Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Ying Sun
- Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Dubo Su
- Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Shun Lv
- Laboratory animal center, Jining Medical University, Jining, 272067, China
| | - Yong Xia
- Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Jing Li
- Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Changlin Li
- Institute of Precision Medicine, Jining Medical University, Jining, 272067, China.
| |
Collapse
|
6
|
Muralidar S, Gopal G, Visaga Ambi S. Targeting the viral-entry facilitators of SARS-CoV-2 as a therapeutic strategy in COVID-19. J Med Virol 2021; 93:5260-5276. [PMID: 33851732 PMCID: PMC8251167 DOI: 10.1002/jmv.27019] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/27/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022]
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19) infection, which has emerged as a global pandemic causing serious concerns. Lack of specific and effective therapeutics for the treatment of COVID-19 is a major concern and the development of vaccines is another important aspect in managing the infection effectively. The first step in the SARS-CoV-2 pathogenesis is the viral entry and it is mediated by its densely glycosylated spike protein (S-protein). Similar to the SARS-CoV, SARS-CoV-2 also engages angiotensin-converting enzyme 2 (ACE2) as the host cell entry receptor. In addition to ACE2, several recent studies have implicated the crucial role of cell surface heparan sulfate (HS) as a necessary assisting cofactor for ACE2-mediated SARS-CoV-2 entry. Furthermore, SARS-CoV-2 was also identified to use both endosomal cysteine proteases cathepsin B and L (CatB/L) and the transmembrane serine protease 2 (TMPRSS2) for the pivotal role of S-protein priming mediating viral entry. As the entry of SARS-CoV-2 into host cells is mandatory for viral infection, it becomes an extremely attractive therapeutic intervention point. In this regard, this review will focus on the therapeutic targeting of the crucial steps of SARS-CoV-2 viral entry like S-protein/ACE2 interaction and S-protein priming by host cell proteases. In addition, this review will also give insights to the readers on several therapeutic opportunities, pharmacological targeting of the viral-entry facilitators like S-Protein, ACE2, cell surface HS, TMPRSS2, and CatB/L and evidence for those drugs currently ongoing clinical studies.
Collapse
Affiliation(s)
- Shibi Muralidar
- Biopharmaceutical Research Lab, Anusandhan Kendra‐1SASTRA Deemed‐to‐be‐UniversityThanjavurTamil NaduIndia
- School of Chemical and BiotechnologySASTRA Deemed‐to‐be‐UniversityThanjavurTamil NaduIndia
| | - Gayathri Gopal
- Biopharmaceutical Research Lab, Anusandhan Kendra‐1SASTRA Deemed‐to‐be‐UniversityThanjavurTamil NaduIndia
- School of Chemical and BiotechnologySASTRA Deemed‐to‐be‐UniversityThanjavurTamil NaduIndia
| | - Senthil Visaga Ambi
- Biopharmaceutical Research Lab, Anusandhan Kendra‐1SASTRA Deemed‐to‐be‐UniversityThanjavurTamil NaduIndia
- School of Chemical and BiotechnologySASTRA Deemed‐to‐be‐UniversityThanjavurTamil NaduIndia
| |
Collapse
|
7
|
Wu AJ, Tong BCK, Huang AS, Li M, Cheung KH. Mitochondrial Calcium Signaling as a Therapeutic Target for Alzheimer's Disease. Curr Alzheimer Res 2021; 17:329-343. [PMID: 31820698 DOI: 10.2174/1567205016666191210091302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 10/17/2019] [Accepted: 12/09/2019] [Indexed: 11/22/2022]
Abstract
Mitochondria absorb calcium (Ca2+) at the expense of the electrochemical gradient generated during respiration. The influx of Ca2+ into the mitochondrial matrix helps maintain metabolic function and results in increased cytosolic Ca2+ during intracellular Ca2+ signaling. Mitochondrial Ca2+ homeostasis is tightly regulated by proteins located in the inner and outer mitochondrial membranes and by the cross-talk with endoplasmic reticulum Ca2+ signals. Increasing evidence indicates that mitochondrial Ca2+ overload is a pathological phenotype associated with Alzheimer's Disease (AD). As intracellular Ca2+ dysregulation can be observed before the appearance of typical pathological hallmarks of AD, it is believed that mitochondrial Ca2+ overload may also play an important role in AD etiology. The high mitochondrial Ca2+ uptake can easily compromise neuronal functions and exacerbate AD progression by impairing mitochondrial respiration, increasing reactive oxygen species formation and inducing apoptosis. Additionally, mitochondrial Ca2+ overload can damage mitochondrial recycling via mitophagy. This review will discuss the molecular players involved in mitochondrial Ca2+ dysregulation and the pharmacotherapies that target this dysregulation. As most of the current AD therapeutics are based on amyloidopathy, tauopathy, and the cholinergic hypothesis, they achieve only symptomatic relief. Thus, determining how to reestablish mitochondrial Ca2+ homeostasis may aid in the development of novel AD therapeutic interventions.
Collapse
Affiliation(s)
- Aston J Wu
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Benjamin C-K Tong
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Alexis S Huang
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| | - King-Ho Cheung
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China.,Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong, China
| |
Collapse
|
8
|
Shukla S, Marks I, Church D, Chan SK, Pokorski JK, Steinmetz NF. Tobacco mosaic virus for the targeted delivery of drugs to cells expressing prostate-specific membrane antigen. RSC Adv 2021; 11:20101-20108. [PMID: 34178308 PMCID: PMC8180379 DOI: 10.1039/d1ra03166j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA) is a membrane-bound protein that is preferentially expressed in the prostate gland and induced in many prostate cancers, making it an important target for new diagnostics and therapeutics. To improve the efficacy of nanoparticle formulations for the imaging and/or eradication of prostate cancer, we synthesized the PSMA-binding glutamic acid derivative DUPA and conjugated it to the external surface of tobacco mosaic virus (TMV) particles. DUPA-targeted TMV was subsequently loaded with the antineoplastic agent mitoxantrone (MTO) or conjugated internally with the fluorescent dye cyanine 5 (Cy5). We found that TMV particles could be efficiently decorated with DUPA and loaded with MTO or Cy5 while maintaining structural integrity. DUPA-targeted TMV particles were able to bind more efficiently to the surface of PSMA+ LNCaP cells compared to non-targeted TMV; but there was little difference in binding efficiency between targeted and untargeted TMV when we tested PSMA− PC3 cells (both cell lines are prostate cancer cell lines). DUPA-targeted TMV particles were internalized by LNCaP cells enabling drug delivery. Finally, we loaded the DUPA-targeted TMV particles and untargeted control particles with MTO to test their cytotoxicity against LNCaP cells in vitro. The cytotoxicity of the TMV-MTO particles (IC50 = 10.2 nM) did not differ significantly from that of soluble MTO at an equivalent dose (IC50 = 12.5 nM) but the targeted particles (TMV-DUPA-MTO) were much more potent (IC50 = 2.80 nM). The threefold increase in cytotoxicity conferred by the DUPA ligand suggests that MTO-loaded, DUPA-coated TMV particles are promising as a therapeutic strategy for PSMA+ prostate cancer and should be advanced to preclinical testing in mouse models of prostate cancer. Prostate-specific membrane antigen (PSMA) is a membrane-bound protein that is preferentially expressed in the prostate gland and induced in many prostate cancers, making it an important target for new diagnostics and therapeutics.![]()
Collapse
Affiliation(s)
- Sourabh Shukla
- Department of NanoEngineering, University of California San Diego La Jolla CA 92093 USA
| | - Isaac Marks
- Department of NanoEngineering, University of California San Diego La Jolla CA 92093 USA
| | - Derek Church
- Department of NanoEngineering, University of California San Diego La Jolla CA 92093 USA
| | - Soo-Khim Chan
- Department of NanoEngineering, University of California San Diego La Jolla CA 92093 USA
| | - Jonathan K Pokorski
- Department of NanoEngineering, University of California San Diego La Jolla CA 92093 USA .,Center for Nano-ImmunoEngineering, University of California San Diego La Jolla CA 92093 USA.,Institute for Materials Discovery and Design, University of California San Diego La Jolla CA 92093 USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego La Jolla CA 92093 USA .,Department of Bioengineering, University of California San Diego La Jolla CA 92093 USA.,Department of Radiology, University of California San Diego La Jolla CA 92093 USA.,Moores Cancer Center, University of California San Diego La Jolla CA 92093 USA.,Center for Nano-ImmunoEngineering, University of California San Diego La Jolla CA 92093 USA.,Institute for Materials Discovery and Design, University of California San Diego La Jolla CA 92093 USA
| |
Collapse
|
9
|
Sharma V, Gupta M, Kumar P, Sharma A. A Comprehensive Review on Fused Heterocyclic as DNA Intercalators: Promising Anticancer Agents. Curr Pharm Des 2021; 27:15-42. [PMID: 33213325 DOI: 10.2174/1381612826666201118113311] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/02/2020] [Indexed: 12/09/2022]
Abstract
Since the discovery of DNA intercalating agents (by Lerman, 1961), a growing number of organic, inorganic, and metallic compounds have been developed to treat life-threatening microbial infections and cancers. Fused-heterocycles are amongst the most important group of compounds that have the ability to interact with DNA. DNA intercalators possess a planar aromatic ring structure that inserts itself between the base pairs of nucleic acids. Once inserted, the aromatic structure makes van der Waals interactions and hydrogen-bonding interactions with the base pairs. The DNA intercalator may also contain an ionizable group that can form ionic interactions with the negatively charged phosphate backbone. After the intercalation, other cellular processes could take place, leading ultimately to cell death. The heterocyclic nucleus present in the DNA intercalators can be considered as a pharmacophore that plays an instrumental role in dictating the affinity and selectivity exhibited by these compounds. In this work, we have carried out a revision of small organic molecules that bind to the DNA molecule via intercalation and cleaving and exert their antitumor activity. A general overview of the most recent results in this area, paying particular attention to compounds that are currently under clinical trials, is provided. Advancement in spectroscopic techniques studying DNA interaction can be examined in-depth, yielding important information on structure-activity relationships. In this comprehensive review, we have focused on the introduction to fused heterocyclic agents with DNA interacting features, from medicinal point of view. The structure-activity relationships points, cytotoxicity data, and binding data and future perspectives of medicinal compounds have been discussed in detail.
Collapse
Affiliation(s)
- Vikas Sharma
- IIMT College of Pharmacy, Knowledge Park III, Greater Noida, Uttar Pradesh-201308, India
| | - Mohit Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Sciences Building, 2730 South Moody Avenue, Portland, OR 97201, United States
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Atul Sharma
- School of Chemistry, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
10
|
Mitoxantrone triggers immunogenic prostate cancer cell death via p53-dependent PERK expression. Cell Oncol (Dordr) 2020; 43:1099-1116. [PMID: 32710433 DOI: 10.1007/s13402-020-00544-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Mitoxantrone (MTX) is a synthetic compound used as a second line chemotherapeutic drug for prostate cancer. It has been reported to trigger immunogenic cell death (ICD) in animal model studies, but the underlying mechanism is not fully understood yet, especially not in prostate cancer cells. METHODS ICD was determined by assessing the release of damage-associated molecular patterns (DAMPs) in the prostate cancer-derived cell lines LNCaP, 22RV1 and PC-3. Short hairpin RNAs (shRNAs) were used to knock down target gene expression. Phagocytosis was assessed using a dual labeling technology in dendric cells co-cultured with cancer cells. The PERK gene promoter was cloned for dual luciferase assays. Chromatin immunoprecipitation (ChIP) was used to determine p53 protein-DNA binding activity. Immunocompetent mice and murine RM-1 prostate cancer cells were used for vaccination experiments. RESULTS MTX treatment induced typical characteristics of DAMP release, including increased cell surface exposure of calreticulin (CALR), and extracellular release of ATP and high mobility group box-1 (HMGB1) protein. MTX also enhanced phagocytosis by dendritic cells. Moreover, MTX treatment increased eukaryotic initiation factor 2α (eIF2α) S51 phosphorylation, which was reduced when PERK and GCN2 were silenced using shRNAs. In addition, PERK or GCN2 silencing significantly reduced MTX-induced release of DAMPs in vitro and anti-tumor immunity in vivo. MTX treatment also resulted in dendritic cell activation in mice, which was attenuated when PERK or GCN2 were silenced in cancer cells used for vaccination. Further analysis revealed that PERK and GCN2 expression was enhanced by MTX treatment, of which PERK, but not GCN2, was enhanced via a p53-dependent mechanism. CONCLUSION MTX triggers ICD by activating eIF2α via PERK/GCN2 upregulation in prostate cancer cells. MTX-induced PERK expression upregulation depends on the p53 pathway, while that of GCN2 requires further investigation.
Collapse
|
11
|
Zhang Q, Chen CZ, Swaroop M, Xu M, Wang L, Lee J, Wang AQ, Pradhan M, Hagen N, Chen L, Shen M, Luo Z, Xu X, Xu Y, Huang W, Zheng W, Ye Y. Heparan sulfate assists SARS-CoV-2 in cell entry and can be targeted by approved drugs in vitro. Cell Discov 2020; 6:80. [PMID: 33298900 PMCID: PMC7610239 DOI: 10.1038/s41421-020-00222-5] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022] Open
Abstract
The cell entry of SARS-CoV-2 has emerged as an attractive drug repurposing target for COVID-19. Here we combine genetics and chemical perturbation to demonstrate that ACE2-mediated entry of SARS-Cov and CoV-2 requires the cell surface heparan sulfate (HS) as an assisting cofactor: ablation of genes involved in HS biosynthesis or incubating cells with a HS mimetic both inhibit Spike-mediated viral entry. We show that heparin/HS binds to Spike directly, and facilitates the attachment of Spike-bearing viral particles to the cell surface to promote viral entry. We screened approved drugs and identified two classes of inhibitors that act via distinct mechanisms to target this entry pathway. Among the drugs characterized, Mitoxantrone is a potent HS inhibitor, while Sunitinib and BNTX disrupt the actin network to indirectly abrogate HS-assisted viral entry. We further show that drugs of the two classes can be combined to generate a synergized activity against SARS-CoV-2-induced cytopathic effect. Altogether, our study establishes HS as an attachment factor that assists SARS coronavirus cell entry and reveals drugs capable of targeting this important step in the viral life cycle.
Collapse
Affiliation(s)
- Qi Zhang
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Catherine Zhengzheng Chen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Manju Swaroop
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Lihui Wang
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Juhyung Lee
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Amy Qiu Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Manisha Pradhan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Natalie Hagen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Lu Chen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Zhiji Luo
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Xin Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Yue Xu
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Wenwei Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
12
|
Mei KC, Liao YP, Jiang J, Chiang M, Khazaieli M, Liu X, Wang X, Liu Q, Chang CH, Zhang X, Li J, Ji Y, Melano B, Telesca D, Xia T, Meng H, Nel AE. Liposomal Delivery of Mitoxantrone and a Cholesteryl Indoximod Prodrug Provides Effective Chemo-immunotherapy in Multiple Solid Tumors. ACS NANO 2020; 14:13343-13366. [PMID: 32940463 PMCID: PMC8023019 DOI: 10.1021/acsnano.0c05194] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
We developed a custom-designed liposome carrier for codelivery of a potent immunogenic cell death (ICD) stimulus plus an inhibitor of the indoleamine 2,3-dioxygenase (IDO-1) pathway to establish a chemo-immunotherapy approach for solid tumors in syngeneic mice. The carrier was constructed by remote import of the anthraquinone chemotherapeutic agent, mitoxantrone (MTO), into the liposomes, which were further endowed with a cholesterol-conjugated indoximod (IND) prodrug in the lipid bilayer. For proof-of-principle testing, we used IV injection of the MTO/IND liposome in a CT26 colon cancer model to demonstrate the generation of a robust immune response, characterized by the appearance of ICD markers (CRT and HMGB-1) as well as evidence of cytotoxic cancer cell death, mediated by perforin and granzyme B. Noteworthy, the cytotoxic effects involved natural killer (NK) cell, which suggests a different type of ICD response. The immunotherapy response was significantly augmented by codelivery of the IND prodrug, which induced additional CRT expression, reduced number of Foxp3+ Treg, and increased perforin release, in addition to extending animal survival beyond the effect of an MTO-only liposome. The outcome reflects the improved pharmacokinetics of MTO delivery to the cancer site by the carrier. In light of the success in the CT26 model, we also assessed the platform efficacy in further breast cancer (EMT6 and 4T1) and renal cancer (RENCA) models, which overexpress IDO-1. Encapsulated MTO delivery was highly effective for inducing chemo-immunotherapy responses, with NK participation, in all tumor models. Moreover, the growth inhibitory effect of MTO was enhanced by IND codelivery in EMT6 and 4T1 tumors. All considered, our data support the use of encapsulated MTO delivery for chemo-immunotherapy, with the possibility to boost the immune response by codelivery of an IDO-1 pathway inhibitor.
Collapse
Affiliation(s)
- Kuo-Ching Mei
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Yu-Pei Liao
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Jinhong Jiang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Michelle Chiang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Mercedeh Khazaieli
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiangsheng Liu
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiang Wang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Qi Liu
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Chong Hyun Chang
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Xiao Zhang
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
| | - Juan Li
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
| | - Ying Ji
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
| | - Brenda Melano
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Donatello Telesca
- Department of Biostatistics, University of California, Los Angeles, California, 90095, United States
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
| | - Huan Meng
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, 90095, United States
| | - Andre E. Nel
- Division of NanoMedicine, Department of Medicine, David Geffen School of Medicine University of California, Los Angeles, California, 90095, United States
- California NanoSystems Institute, University of California, Los Angeles, California 90095, United States
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, 90095, United States
| |
Collapse
|
13
|
El-Helby AGA, Sakr H, Ayyad RR, Mahdy HA, Khalifa MM, Belal A, Rashed M, El-Sharkawy A, Metwaly AM, Elhendawy MA, Radwan MM, ElSohly MA, Eissa IH. Design, synthesis, molecular modeling, in vivo studies and anticancer activity evaluation of new phthalazine derivatives as potential DNA intercalators and topoisomerase II inhibitors. Bioorg Chem 2020; 103:104233. [DOI: 10.1016/j.bioorg.2020.104233] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 12/17/2022]
|
14
|
Zhang Q, Chen CZ, Swaroop M, Xu M, Wang L, Lee J, Wang AQ, Pradhan M, Hagen N, Chen L, Shen M, Luo Z, Xu X, Xu Y, Huang W, Zheng W, Ye Y. Heparan sulfate assists SARS-CoV-2 in cell entry and can be targeted by approved drugs in vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.14.202549. [PMID: 32699847 PMCID: PMC7373127 DOI: 10.1101/2020.07.14.202549] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The cell entry of SARS-CoV-2 has emerged as an attractive drug repurposing target for COVID-19. Here we combine genetics and chemical perturbation to demonstrate that ACE2-mediated entry of SARS-CoV and CoV-2 requires the cell surface heparan sulfate (HS) as an assisting cofactor: ablation of genes involved in HS biosynthesis or incubating cells with a HS mimetic both inhibit Spike-mediated viral entry. We show that heparin/HS binds to Spike directly, facilitates the attachment of viral particles to the cell surface to promote cell entry. We screened approved drugs and identified two classes of inhibitors that act via distinct mechanisms to target this entry pathway. Among the drugs characterized, Mitoxantrone is a potent HS inhibitor, while Sunitinib and BNTX disrupt the actin network to indirectly abrogate HS-assisted viral entry. We further show that drugs of the two classes can be combined to generate a synergized activity against SARS-CoV-2-induced cytopathic effect. Altogether, our study establishes HS as an attachment factor that assists SARS coronavirus cell entry, and reveals drugs capable of targeting this important step in the viral life cycle.
Collapse
Affiliation(s)
- Qi Zhang
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Catherine Z. Chen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Manju Swaroop
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Lihui Wang
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Juhyung Lee
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Amy Q. Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Manisha Pradhan
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Natalie Hagen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Lu Chen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Zhiji Luo
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Xin Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Yue Xu
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Wenwei Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
15
|
Ionescu A, Caligiuri R, Godbert N, Ricciardi L, La Deda M, Ghedini M, Ferri N, Lupo MG, Facchetti G, Rimoldi I, Aiello I. Cytotoxic performances of new anionic cyclometalated Pt(II) complexes bearing chelated O^O ligands. Appl Organomet Chem 2020. [DOI: 10.1002/aoc.5455] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Andreea Ionescu
- MAT‐InLAB, LASCAMM CR‐INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie ChimicheUniversità della Calabria Ponte Pietro Bucci Cubo 14C 87036 Arcavacata di Rende (CS) Italy
- CNR NANOTEC‐Istituto di Nanotecnologia UOS Cosenza 87036 Arcavacata di Rende (CS) Italy
| | - Rossella Caligiuri
- MAT‐InLAB, LASCAMM CR‐INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie ChimicheUniversità della Calabria Ponte Pietro Bucci Cubo 14C 87036 Arcavacata di Rende (CS) Italy
| | - Nicolas Godbert
- MAT‐InLAB, LASCAMM CR‐INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie ChimicheUniversità della Calabria Ponte Pietro Bucci Cubo 14C 87036 Arcavacata di Rende (CS) Italy
- CNR NANOTEC‐Istituto di Nanotecnologia UOS Cosenza 87036 Arcavacata di Rende (CS) Italy
| | - Loredana Ricciardi
- CNR NANOTEC‐Istituto di Nanotecnologia UOS Cosenza 87036 Arcavacata di Rende (CS) Italy
| | - Massimo La Deda
- MAT‐InLAB, LASCAMM CR‐INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie ChimicheUniversità della Calabria Ponte Pietro Bucci Cubo 14C 87036 Arcavacata di Rende (CS) Italy
- CNR NANOTEC‐Istituto di Nanotecnologia UOS Cosenza 87036 Arcavacata di Rende (CS) Italy
| | - Mauro Ghedini
- MAT‐InLAB, LASCAMM CR‐INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie ChimicheUniversità della Calabria Ponte Pietro Bucci Cubo 14C 87036 Arcavacata di Rende (CS) Italy
- CNR NANOTEC‐Istituto di Nanotecnologia UOS Cosenza 87036 Arcavacata di Rende (CS) Italy
| | - Nicola Ferri
- Dipartimento di Scienze del FarmacoUniversità degli Studi di Padova Via Marzolo 5 35131 Padua Italy
| | - Maria Giovanna Lupo
- Dipartimento di Scienze del FarmacoUniversità degli Studi di Padova Via Marzolo 5 35131 Padua Italy
| | - Giorgio Facchetti
- Dipartimento di Scienze FarmaceuticheUniversità degli Studi di Milano Via Venezian 21 20133 Milan Italy
| | - Isabella Rimoldi
- Dipartimento di Scienze FarmaceuticheUniversità degli Studi di Milano Via Venezian 21 20133 Milan Italy
| | - Iolinda Aiello
- MAT‐InLAB, LASCAMM CR‐INSTM, Unità INSTM della Calabria, Dipartimento di Chimica e Tecnologie ChimicheUniversità della Calabria Ponte Pietro Bucci Cubo 14C 87036 Arcavacata di Rende (CS) Italy
- CNR NANOTEC‐Istituto di Nanotecnologia UOS Cosenza 87036 Arcavacata di Rende (CS) Italy
| |
Collapse
|
16
|
Padmapriya K, Barthwal R. Nuclear magnetic resonance based structure of the protoberberine alkaloid coralyne and its self-association by spectroscopy techniques. J Pharm Anal 2019; 9:437-448. [PMID: 31890344 PMCID: PMC6931075 DOI: 10.1016/j.jpha.2019.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 08/19/2019] [Accepted: 09/26/2019] [Indexed: 12/03/2022] Open
Abstract
Coralyne is an important alkaloid due to its anti-cancer and other medicinal properties. It targets DNA in cells and acts as human topoisomerase-I poison, telomerase inhibitor and nucleic acid intercalator. It has high tendency to undergo self-association, which is a matter of concern for therapeutic applications. The understanding of its interaction with DNA requires precise knowledge of chemical shifts in Nuclear Magnetic Resonance (NMR) spectra besides self-association. The present study is the first report of a complete assignment of all 1H/13C resonances in NMR spectra of coralyne in DMSO-d6 using one dimensional 1H/13C and two dimensional NMR experiments. The chemical shift of all proton and several 13C resonances have also been obtained in D2O and ethanol-d6. The same has been calculated using Density Functional Theory (DFT). NMR spectra of coralyne show upfield shift of 0.6–1.2 ppm in aromatic ring protons suggesting stacking interactions. Apart from 11 intra molecular NOE cross peaks in 2D 1H–1H ROESY spectra, 3 short distance NOE correlations, H6–10OCH3, H5–10OCH3 and H12–16CH3, give direct independent evidence of the formation of a stacked dimer. The absorbance, fluorescence, circular dichroism and fluorescence lifetime experiments conducted in the present investigations corroborate results obtained by NMR. First report of NMR chemical shifts of all 1H and 13C resonances in coralyne. Proton and carbon-13 chemical shifts calculated using Density Functional theory. Self associated coralyne shows upfield shifts up to ∼1.2 ppm in proton resonances. Three intermolecular NOEs in 2D ROESY spectra give direct proof of dimer formation. Absorbance, fluorescence and life time experiments give evidence of dimer formation.
Collapse
Affiliation(s)
- Kumar Padmapriya
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, India
| | - Ritu Barthwal
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, India
| |
Collapse
|
17
|
Woods J, Nemani N, Shanmughapriya S, Kumar A, Zhang M, Nathan SR, Thomas M, Carvalho E, Ramachandran K, Srikantan S, Stathopulos PB, Wilson JJ, Madesh M. A Selective and Cell-Permeable Mitochondrial Calcium Uniporter (MCU) Inhibitor Preserves Mitochondrial Bioenergetics after Hypoxia/Reoxygenation Injury. ACS CENTRAL SCIENCE 2019; 5:153-166. [PMID: 30693334 PMCID: PMC6346394 DOI: 10.1021/acscentsci.8b00773] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Indexed: 05/10/2023]
Abstract
Mitochondrial Ca2+ (mCa2+) uptake mediated by the mitochondrial calcium uniporter (MCU) plays a critical role in signal transduction, bioenergetics, and cell death, and its dysregulation is linked to several human diseases. In this study, we report a new ruthenium complex Ru265 that is cell-permeable, minimally toxic, and highly potent with respect to MCU inhibition. Cells treated with Ru265 show inhibited MCU activity without any effect on cytosolic Ca2+ dynamics and mitochondrial membrane potential (ΔΨm). Dose-dependent studies reveal that Ru265 is more potent than the currently employed MCU inhibitor Ru360. Site-directed mutagenesis of Cys97 in the N-terminal domain of human MCU ablates the inhibitory activity of Ru265, suggesting that this matrix-residing domain is its target site. Additionally, Ru265 prevented hypoxia/reoxygenation injury and subsequent mitochondrial dysfunction, demonstrating that this new inhibitor is a valuable tool for studying the functional role of the MCU in intact biological models.
Collapse
Affiliation(s)
- Joshua
J. Woods
- Robert
F. Smith School for Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853, United States
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United
States
| | - Neeharika Nemani
- Department
of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
for Translational Medicine, Lewis Katz School
of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Santhanam Shanmughapriya
- Department
of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
for Translational Medicine, Lewis Katz School
of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Akshay Kumar
- Department
of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
for Translational Medicine, Lewis Katz School
of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | - MengQi Zhang
- Department
of Physiology and Pharmacology, Western
University, London, Ontario N6A 5C1, Canada
| | - Sarah R. Nathan
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United
States
| | - Manfred Thomas
- Department
of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
for Translational Medicine, Lewis Katz School
of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Edmund Carvalho
- Department
of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
for Translational Medicine, Lewis Katz School
of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Karthik Ramachandran
- Department
of Medicine/Nephrology, Institute for Precision Medicine and Health, University of Texas Health San Antonio, San Antonio, Texas 78229, United States
| | - Subramanya Srikantan
- Department
of Medicine/Nephrology, Institute for Precision Medicine and Health, University of Texas Health San Antonio, San Antonio, Texas 78229, United States
| | - Peter B. Stathopulos
- Department
of Physiology and Pharmacology, Western
University, London, Ontario N6A 5C1, Canada
| | - Justin J. Wilson
- Department
of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York 14853, United
States
| | - Muniswamy Madesh
- Department
of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
- Center
for Translational Medicine, Lewis Katz School
of Medicine at Temple University, Philadelphia, Pennsylvania 19140, United States
- Department
of Medicine/Nephrology, Institute for Precision Medicine and Health, University of Texas Health San Antonio, San Antonio, Texas 78229, United States
| |
Collapse
|
18
|
Halicka HD, Li J, Zhao H, Darzynkiewicz Z. Concurrent detection of lysosome and tissue transglutaminase activation in relation to cell cycle position during apoptosis induced by different anticancer drugs. Cytometry A 2018; 95:683-690. [PMID: 30422397 DOI: 10.1002/cyto.a.23652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/18/2018] [Accepted: 09/28/2018] [Indexed: 12/16/2022]
Abstract
Described is the new cytometric approach do detect either stimulation or a collapse of lysosomal proton pump (lysosomes rupture) combined with activation of transglutaminase 2 (TG2) during induction of apoptosis. Apoptosis of human lymphoblastoid TK6 cells was induced by combination of 2-deoxyglucose with the isoquinoline alkaloid berberine, by DNA topoisomerase I inhibitor camptothecin, its analog topotecan, topoisomerase II inhibitors etoposide or mitoxantrone, as well as by the cytotoxic anticancer ribonuclease ranpirnase (onconase). Activity of the proton pump of lysosomes was assessed by measuring entrapment and accumulation of the basic fluorochrome acridine orange (AO) resulting in its metachromatic red luminescence (F>640 ) within these organelles. Activation of TG2 was detected in the same cell subpopulation by the evidence of crosslinking of cytoplasmic proteins revealed by the increased intensity of the side light scatter (SSC) as well as following cell lysis by detergent, by its red fluorescence after staining by sulforhodamine 101. Because at low AO concentration nuclear DNA of the lysed cells was stoichiometrically stained green (F530 ) its quantity provided information on effects of the drug treatments on cell cycle in relation to activation of TG2. The data reveal that activation of lysosomal proton pump was evident in subpopulations of cells treated with 2-deoxyglucose plus berberine, topotecan, etoposide and mitoxantrone but not with ranpirnase. The collapse of lysosomal proton pump possibly reporting rupture of these organelles was observed in definite cell subpopulations after treatment with each of the studied drugs. Because regardless of the inducer of apoptosis TG2 activation invariably was correlated with lysosomes rupture it is likely that it was triggered by calcium ions or protons released from the ruptured lysosomes. This new methodological approach offers the means to investigate mechanisms and factors affecting autophagic lysosomes proton pump activity vis-à-vis TG2 activation that are common in several pathological states. © 2019 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- H Dorota Halicka
- Department of Pathology, New York Medical College, Brander Cancer Research Institute, Valhalla, New York
| | - Jiangwei Li
- Department of Pathology, New York Medical College, Brander Cancer Research Institute, Valhalla, New York
| | - Hong Zhao
- Department of Pathology, New York Medical College, Brander Cancer Research Institute, Valhalla, New York
| | - Zbigniew Darzynkiewicz
- Department of Pathology, New York Medical College, Brander Cancer Research Institute, Valhalla, New York
| |
Collapse
|
19
|
Deo KM, Ang DL, McGhie B, Rajamanickam A, Dhiman A, Khoury A, Holland J, Bjelosevic A, Pages B, Gordon C, Aldrich-Wright JR. Platinum coordination compounds with potent anticancer activity. Coord Chem Rev 2018. [DOI: 10.1016/j.ccr.2017.11.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
20
|
Tamura RE, de Luna IV, Lana MG, Strauss BE. Improving adenoviral vectors and strategies for prostate cancer gene therapy. Clinics (Sao Paulo) 2018; 73:e476s. [PMID: 30133562 PMCID: PMC6097088 DOI: 10.6061/clinics/2018/e476s] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/09/2018] [Indexed: 01/07/2023] Open
Abstract
Gene therapy has been evaluated for the treatment of prostate cancer and includes the application of adenoviral vectors encoding a suicide gene or oncolytic adenoviruses that may be armed with a functional transgene. In parallel, versions of adenoviral vector expressing the p53 gene (Ad-p53) have been tested as treatments for head and neck squamous cell carcinoma and non-small cell lung cancer. Although Ad-p53 gene therapy has yielded some interesting results when applied to prostate cancer, it has not been widely explored, perhaps due to current limitations of the approach. To achieve better functionality, improvements in the gene transfer system and the therapeutic regimen may be required. We have developed adenoviral vectors whose transgene expression is controlled by a p53-responsive promoter, which creates a positive feedback mechanism when used to drive the expression of p53. Together with improvements that permit efficient transduction, this new approach was more effective than the use of traditional versions of Ad-p53 in killing prostate cancer cell lines and inhibiting tumor progression. Even so, gene therapy is not expected to replace traditional chemotherapy but should complement the standard of care. In fact, chemotherapy has been shown to assist in viral transduction and transgene expression. The cooperation between gene therapy and chemotherapy is expected to effectively kill tumor cells while permitting the use of reduced chemotherapy drug concentrations and, thus, lowering side effects. Therefore, the combination of gene therapy and chemotherapy may prove essential for the success of both approaches.
Collapse
Affiliation(s)
- Rodrigo Esaki Tamura
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Igor Vieira de Luna
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Marlous Gomes Lana
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Bryan E Strauss
- Laboratório de Vetores Virais, Centro de Investigação Translacional em Oncologia, Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail: ,
| |
Collapse
|
21
|
Suresh N, Suresh A, Yerramsetty S, Bhadra MP, Alvala M, Sekhar KVGC. Anti-proliferative activity, molecular modeling studies and interaction with calf thymus DNA of novel ciprofloxacin analogues. J CHEM SCI 2018. [DOI: 10.1007/s12039-018-1528-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Kreft D, Wang Y, Rattay M, Toensing K, Anselmetti D. Binding mechanism of anti-cancer chemotherapeutic drug mitoxantrone to DNA characterized by magnetic tweezers. J Nanobiotechnology 2018; 16:56. [PMID: 30005668 PMCID: PMC6043947 DOI: 10.1186/s12951-018-0381-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/26/2018] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Chemotherapeutic agents (anti-cancer drugs) are small cytostatic or cytotoxic molecules that often bind to double-stranded DNA (dsDNA) resulting in modifications of their structural and nanomechanical properties and thus interfering with the cell proliferation process. METHODS We investigated the anthraquinone compound mitoxantrone that is used for treating certain cancer types like leukemia and lymphoma with magnetic tweezers as a single molecule nanosensor. In order to study the association of mitoxantrone with dsDNA, we conducted force-extension and mechanical overwinding experiments with a sensitivity of 10-14 N. RESULTS Using this method, we were able to estimate an equilibrium constant of association Ka ≈ 1 × 105 M-1 as well as a binding site size of n ≈ 2.5 base pairs for mitoxantrone. An unwinding angle of mitoxantrone-intercalation of ϑ ≈ 16° was determined. CONCLUSION Moreover, we observed a complex concentration-dependent bimodal binding behavior, where mitoxantrone associates to dsDNA as an intercalator and groove binder simultaneously at low concentrations and as a mere intercalator at high concentrations.
Collapse
Affiliation(s)
- Dennis Kreft
- Experimental Biophysics and Applied Nanoscience, Physics Department, Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Universitaetsstrasse 25, 33615 Bielefeld, Germany
| | - Ying Wang
- Experimental Biophysics and Applied Nanoscience, Physics Department, Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Universitaetsstrasse 25, 33615 Bielefeld, Germany
| | - Michael Rattay
- Baxter Oncology GmbH, Kantstrasse 2, 33790 Halle Westphalia, Germany
| | - Katja Toensing
- Experimental Biophysics and Applied Nanoscience, Physics Department, Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Universitaetsstrasse 25, 33615 Bielefeld, Germany
| | - Dario Anselmetti
- Experimental Biophysics and Applied Nanoscience, Physics Department, Bielefeld Institute for Nanoscience (BINAS), Bielefeld University, Universitaetsstrasse 25, 33615 Bielefeld, Germany
| |
Collapse
|
23
|
Pawlik A, Szczepanski MA, Klimaszewska-Wisniewska A, Gackowska L, Zuryn A, Grzanka A. Cytoskeletal reorganization and cell death in mitoxantrone-treated lung cancer cells. Acta Histochem 2016; 118:784-796. [PMID: 27817864 DOI: 10.1016/j.acthis.2016.10.001] [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] [Received: 05/23/2016] [Accepted: 10/14/2016] [Indexed: 10/20/2022]
Abstract
The aim of this study was to investigate the cytotoxic effect of mitoxantrone on two human non-small cell lung cancer cell lines, A549 (p53+) and H1299 (p53-). To our knowledge, this is the first study to evaluate the impact of MXT on the organization of cytoskeletal proteins. Analyses were performed using fluorescence and transmission electron microscopy, spectrophotometric techniques, flow cytometry and Western blotting. It was shown that H1299 cells are significantly more sensitive to mitoxantrone than the A549 cell line, and that the growth-inhibitory effect of the drug is dose-dependent only after longer incubation. The observed presence of ring-like microtubule structures and mitochondria surrounding the nuclei of H1299 cells could be a manifestation of increased tubulin polymerization requiring large amounts of energy, whereas the loss of actin stress fibers was presumably not the cause but rather the consequence of cell death induction. Treatment with mitoxantrone also led to the appearance of structures resembling agresomes in H1299 cells and to nucleolar segregation in both cell lines. It was demonstrated that cells arrested in the S phase were most susceptible to cell death induction, and that triggered intracellular changes led mainly to apoptosis. High concentrations induced necrosis and some H1299 cells exhibited morphological features of mitotic catastrophe.
Collapse
|
24
|
Singla P, Luxami V, Singh R, Tandon V, Paul K. Novel pyrazolo[3,4-d]pyrimidine with 4-(1H-benzimidazol-2-yl)-phenylamine as broad spectrum anticancer agents: Synthesis, cell based assay, topoisomerase inhibition, DNA intercalation and bovine serum albumin studies. Eur J Med Chem 2016; 126:24-35. [PMID: 27744184 DOI: 10.1016/j.ejmech.2016.09.093] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/03/2016] [Accepted: 09/28/2016] [Indexed: 12/19/2022]
Abstract
A series of new pyrazolo[3,4-d]pyrimidine possessing 4-(1H-benzimidazol-2-yl)-phenylamine moiety at C4 position and primary as well as secondary amines at C6 position has been designed and synthesized. Their antitumor activities were evaluated against a panel of 60 human cancer cell lines at National Cancer Institute (NCI). Six compounds displayed potent and broad spectrum anticancer activities at 10 μM. Compounds 8, 12, 14 and 17 proved to be the most active and efficacious candidate in this series, with mean GI50 values of 1.30 μM, 1.43 μM, 2.38 μM and 2.18 μM, respectively against several cancer cell lines. Further biological evaluation of these compounds suggested that these compounds induce apoptosis and inhibit human topoisomerase (Topo) IIα as a possible intracellular target. UV-visible and fluorescence studies of these compounds revealed strong interaction with ct-DNA and bovine serum albumin (BSA).
Collapse
Affiliation(s)
- Prinka Singla
- School of Chemistry and Biochemistry, Thapar University, Patiala, 147004, India
| | - Vijay Luxami
- School of Chemistry and Biochemistry, Thapar University, Patiala, 147004, India.
| | - Raja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110 067, India
| | - Vibha Tandon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110 067, India
| | - Kamaldeep Paul
- School of Chemistry and Biochemistry, Thapar University, Patiala, 147004, India.
| |
Collapse
|
25
|
Pradeep TP, Barthwal R. A 4:1 stoichiometric binding and stabilization of mitoxantrone-parallel stranded G-quadruplex complex established by spectroscopy techniques. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2016; 162:106-114. [DOI: 10.1016/j.jphotobiol.2016.06.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 06/10/2016] [Accepted: 06/11/2016] [Indexed: 12/30/2022]
|
26
|
Pradeep TP, Barthwal R. NMR structure of dual site binding of mitoxantrone dimer to opposite grooves of parallel stranded G-quadruplex [d-(TTGGGGT)]4. Biochimie 2016; 128-129:59-69. [DOI: 10.1016/j.biochi.2016.07.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 07/11/2016] [Indexed: 12/27/2022]
|
27
|
de Campos-Nebel M, Palmitelli M, González-Cid M. A flow cytometry-based method for a high-throughput analysis of drug-stabilized topoisomerase II cleavage complexes in human cells. Cytometry A 2016; 89:852-60. [PMID: 27517472 DOI: 10.1002/cyto.a.22919] [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: 04/15/2016] [Revised: 06/27/2016] [Accepted: 07/15/2016] [Indexed: 11/08/2022]
Abstract
Topoisomerase II (Top2) is an important target for anticancer therapy. A variety of drugs that poison Top2, including several epipodophyllotoxins, anthracyclines, and anthracenediones, are widely used in the clinic for both hematologic and solid tumors. The poisoning of Top2 involves the formation of a reaction intermediate Top2-DNA, termed Top2 cleavage complex (Top2cc), which is persistent in the presence of the drug and involves a 5' end of DNA covalently bound to a tyrosine from the enzyme through a phosphodiester group. Drug-induced Top2cc leads to Top2 linked-DNA breaks which are the major responsible for their cytotoxicity. While biochemical detection is very laborious, quantification of drug-induced Top2cc by immunofluorescence-based microscopy techniques is time consuming and requires extensive image segmentation for the analysis of a small population of cells. Here, we developed a flow cytometry-based method for the analysis of drug-induced Top2cc. This method allows a rapid analysis of a high number of cells in their cell cycle phase context. Moreover, it can be applied to almost any human cell type, including clinical samples. The methodology is useful for a high-throughput analysis of drugs that poison Top2, allowing not just the discrimination of the Top2 isoform that is targeted but also to track its removal. © 2016 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Marcelo de Campos-Nebel
- Laboratorio de Mutagénesis, Instituto de Medicina Experimental (IMEX), Academia Nacional de Medicina, CONICET, Buenos Aires, Argentina.
| | - Micaela Palmitelli
- Laboratorio de Mutagénesis, Instituto de Medicina Experimental (IMEX), Academia Nacional de Medicina, CONICET, Buenos Aires, Argentina
| | - Marcela González-Cid
- Laboratorio de Mutagénesis, Instituto de Medicina Experimental (IMEX), Academia Nacional de Medicina, CONICET, Buenos Aires, Argentina
| |
Collapse
|
28
|
Jamison JM, Krabill K, Allen KA, Stuart SH, Tsai CC. RNA–Intercalating Agent Interactions: in vitro Antiviral Activity Studies. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029000100601] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Twenty intercalating agents were tested to examine the effects of intercalating dye-induced perturbations upon the antiviral activity of poly (adenylate–uridylate) [poly (A-U)]. Neither poly (A-U) alone nor each intercalative dye was an efficacious antiviral agent. When poly (A-U) was combined with major groove intercalating dyes (acridine orange or proflavine), no synergism was observed. When poly (A-U) was combined with minor groove intercalating dyes [ethidium (EB), propidium (PI), adriamycin (ADR) or daunomycin (DMN)] or minor/major groove intercalating dyes [9-aminoacridine (9-AA), N2-methyl-9-hydroxy-ellipticine (NMHE) or N2,N6-dimethyl-9-hydroxy-ellipticine (DMHE)] the 50% effective doses (ED50) of the poly (A-U), 9-AA, ADR, DMHE, DMN, EB, NMHE and PI decreased 18-, 22-, 60-, 274-, 61-, 154-, 113- and 299-fold, respectively. When poly (A-U) was combined individually with 11 dyes whose mode of intercalation was not known, the ED50 of ametantrone (HAQ), chloroquine (CHL), mitoxantrone (DHAQ) and quinine (QUI) decreased 125-, 65-, 251- and 32-fold, respectively. These results suggest that the four dyes may intercalate into poly (A-U) from the minor groove. Ten (ADR, CHL, DMN, DHAQ, DMHE, EB, HAQ, NMHE, PI, QUI) of the 20 dyes evaluated exhibited significant synergism with poly (A-U), as quantified by the fractional inhibitory concentration index. Interferon (IFN) neutralization assays demonstrated that the IFN-inducing capability of the dye/poly (A-U) combinations approximated the sum of the capabilities of the poly (A-U) and the dyes employed. These results suggest that the majority of the dyes tested potentiate the antiviral activity of poly (A-U) without affecting the amount of IFN induced.
Collapse
Affiliation(s)
- J. M. Jamison
- Department of Chemistry, Kent State University, Kent, OH 44242, USA
- Department of Microbiology and Immunology, Northeastern Ohio Universities College of Medicine, Rootstown, OH 44272, USA
| | - K. Krabill
- Department of Chemistry, Kent State University, Kent, OH 44242, USA
- Department of Microbiology and Immunology, Northeastern Ohio Universities College of Medicine, Rootstown, OH 44272, USA
| | - K. A. Allen
- Department of Microbiology and Immunology, Northeastern Ohio Universities College of Medicine, Rootstown, OH 44272, USA
| | - S. H. Stuart
- Department of Microbiology and Immunology, Northeastern Ohio Universities College of Medicine, Rootstown, OH 44272, USA
| | - C.-C. Tsai
- Department of Chemistry, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
29
|
Narva S, Chitti S, Bala BR, Alvala M, Jain N, Kondapalli VGCS. Synthesis and biological evaluation of pyrrolo[2,3- b ]pyridine analogues as antiproliferative agents and their interaction with calf thymus DNA. Eur J Med Chem 2016; 114:220-31. [DOI: 10.1016/j.ejmech.2016.02.059] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/23/2016] [Accepted: 02/24/2016] [Indexed: 12/11/2022]
|
30
|
Pentoxifylline affects idarubicin binding to DNA. Bioorg Chem 2016; 65:118-25. [DOI: 10.1016/j.bioorg.2016.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 01/21/2023]
|
31
|
Meneghello M, Papadopoulou E, Ugo P, Bartlett PN. Using Electrochemical SERS to Measure the Redox Potential of Drug Molecules Bound to dsDNA—a Study of Mitoxantrone. Electrochim Acta 2016. [DOI: 10.1016/j.electacta.2015.11.121] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
32
|
Gołuński G, Borowik A, Wyrzykowski D, Woziwodzka A, Piosik J. Pentoxifylline as a modulator of anticancer drug doxorubicin. Part I: Reduction of doxorubicin DNA binding. Chem Biol Interact 2015; 242:291-8. [PMID: 26499448 DOI: 10.1016/j.cbi.2015.10.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 10/07/2015] [Accepted: 10/08/2015] [Indexed: 12/26/2022]
Abstract
Pentoxifylline--biologically active aromatic compound--has a well established capability to sequester aromatic ligands, such as an anticancer drug--doxorubicin--in mixed stacking aggregates. Formation of such hetero-complexes may influence biological activity of secluded drug. Presented work shows assessment of pentoxifylline influence on doxorubicin direct interactions with DNA employing biophysical methods. Achievement of this goal required statistical-thermodynamical model allowing numerical four-parameter analysis of experimental mixture--an issue that was successfully tackled by merging McGhee--von Hippel and Kapuscinski--Kimmel models. Results obtained with new model are well in agreement with data obtained with separate experiments with each of these two models and show reduction of doxorubicin in free (monomeric, dimeric) and complexed with DNA forms in favor of doxorubicin-pentoxifylline complexes with increasing pentoxifylline concentration. Developed model appears to be a universal tool allowing numerical analysis of mixtures containing self-aggregating ligand, DNA, and modulating agent.
Collapse
Affiliation(s)
- Grzegorz Gołuński
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology UG-MUG, Kładki 24, 80-822, Gdańsk, Poland
| | - Agnieszka Borowik
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology UG-MUG, Kładki 24, 80-822, Gdańsk, Poland
| | - Dariusz Wyrzykowski
- Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, 80-308, Gdańsk, Poland
| | - Anna Woziwodzka
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology UG-MUG, Kładki 24, 80-822, Gdańsk, Poland.
| | - Jacek Piosik
- Laboratory of Biophysics, Intercollegiate Faculty of Biotechnology UG-MUG, Kładki 24, 80-822, Gdańsk, Poland.
| |
Collapse
|
33
|
Evison BJ, Sleebs BE, Watson KG, Phillips DR, Cutts SM. Mitoxantrone, More than Just Another Topoisomerase II Poison. Med Res Rev 2015; 36:248-99. [PMID: 26286294 DOI: 10.1002/med.21364] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 02/06/2023]
Abstract
Mitoxantrone is a synthetic anthracenedione originally developed to improve the therapeutic profile of the anthracyclines and is commonly applied in the treatment of breast and prostate cancers, lymphomas, and leukemias. A comprehensive overview of the drug's molecular, biochemical, and cellular pharmacology is presented here, beginning with the cardiotoxic nature of its predecessor doxorubicin and how these properties shaped the pharmacology of mitoxantrone itself. Although mitoxantrone is firmly established as a DNA topoisomerase II poison within mammalian cells, it is now clear that the drug interacts with a much broader range of biological macromolecules both covalently and noncovalently. Here, we consider each of these interactions in the context of their wider biological relevance to cancer therapy and highlight how they may be exploited to further enhance the therapeutic value of mitoxantrone. In doing so, it is now clear that mitoxantrone is more than just another topoisomerase II poison.
Collapse
Affiliation(s)
- Benny J Evison
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| | - Brad E Sleebs
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Keith G Watson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3052, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Don R Phillips
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| | - Suzanne M Cutts
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Victoria, 3086, Australia
| |
Collapse
|
34
|
Shaul P, Steinbuch KB, Blacher E, Stein R, Fridman M. Exploring the Effects of Glycosylation and Etherification of the Side Chains of the Anticancer Drug Mitoxantrone. ChemMedChem 2015; 10:1528-38. [DOI: 10.1002/cmdc.201500274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 02/02/2023]
|
35
|
Probing the interactions of mitoxantrone with biomimetic membranes with electrochemical and spectroscopic techniques. Electrochim Acta 2015. [DOI: 10.1016/j.electacta.2015.02.223] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Zhao H, Darzynkiewicz Z. Attenuation of replication stress-induced premature cellular senescence to assess anti-aging modalities. ACTA ACUST UNITED AC 2014; 69:9.47.1-9.47.10. [PMID: 24984966 DOI: 10.1002/0471142956.cy0947s69] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Described is an in vitro model of premature senescence in pulmonary adenocarcinoma A549 cells induced by persistent DNA replication stress in response to treatment with the DNA damaging drug mitoxantrone (Mxt). The degree of cellular senescence, based on characteristic changes in cell morphology, is measured by laser scanning cytometry. Specifically, the flattening of cells grown on slides (considered the hallmark of cellular senescence) is measured as the decline in local intensity of DNA-associated DAPI fluorescence (represented by maximal pixels). This change is paralleled by an increase in nuclear area. Thus, the ratio of mean intensity of maximal pixels to nuclear area provides a very sensitive morphometric biomarker for the degree of senescence. This analysis is combined with immunocytochemical detection of senescence markers, such as overexpression of cyclin kinase inhibitors (e.g., p21(WAF1) ) and phosphorylation of ribosomal protein S6 (rpS6), a key marker associated with aging/senescence that is detected using a phospho-specific antibody. These biomarker indices are presented in quantitative terms defined as a senescence index (SI), which is the fraction of the marker in test cultures relative to the same marker in exponentially growing control cultures. This system can be used to evaluate the anti-aging potential of test agents by assessing attenuation of maximal senescence. As an example, the inclusion of berberine, a natural alkaloid with reported anti-aging properties and a long history of use in traditional Chinese medicine, is shown to markedly attenuate the Mxt-induced SI and phosphorylation of rpS6. The multivariate analysis of senescence markers by laser scanning cytometry offers a promising tool to explore the potential anti-aging properties of a variety agents.
Collapse
Affiliation(s)
- Hong Zhao
- Brander Cancer Research Institute and Department of Pathology, New York Medical College, Valhalla, New York
| | | |
Collapse
|
37
|
Wu CC, Li YC, Wang YR, Li TK, Chan NL. On the structural basis and design guidelines for type II topoisomerase-targeting anticancer drugs. Nucleic Acids Res 2013; 41:10630-40. [PMID: 24038465 PMCID: PMC3905874 DOI: 10.1093/nar/gkt828] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Type II topoisomerases (Top2s) alter DNA topology via the formation of an enzyme-DNA adduct termed cleavage complex, which harbors a transient double-strand break in one DNA to allow the passage of another. Agents targeting human Top2s are clinically active anticancer drugs whose trapping of Top2-mediated DNA breakage effectively induces genome fragmentation and cell death. To understand the structural basis of this drug action, we previously determined the structure of human Top2 β-isoform forming a cleavage complex with the drug etoposide and DNA, and described the insertion of drug into DNA cleavage site and drug-induced decoupling of catalytic groups. By developing a post-crystallization drug replacement procedure that simplifies structural characterization of drug-stabilized cleavage complexes, we have extended the analysis toward other structurally distinct drugs, m-AMSA and mitoxantrone. Besides the expected drug intercalation, a switch in ribose puckering in the 3'-nucleotide of the cleavage site was robustly observed in the new structures, representing a new mechanism for trapping the Top2 cleavage complex. Analysis of drug-binding modes and the conformational landscapes of the drug-binding pockets provide rationalization of the drugs' structural-activity relationships and explain why Top2 mutants exhibit differential effects toward each drug. Drug design guidelines were proposed to facilitate the development of isoform-specific Top2-targeting anticancer agents.
Collapse
Affiliation(s)
- Chyuan-Chuan Wu
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan, Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and Center for Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | - Yi-Ching Li
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan, Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and Center for Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | - Ying-Ren Wang
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan, Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and Center for Biotechnology, National Taiwan University, Taipei 106, Taiwan
| | - Tsai-Kun Li
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan, Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and Center for Biotechnology, National Taiwan University, Taipei 106, Taiwan,Correspondence may also be addressed to Tsai-Kun Li. Tel: +886 2 22123456 (ext 88287/88294); Fax: +886 2 23915293;
| | - Nei-Li Chan
- Institute of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan, Institute of Biochemistry, College of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan, Department and Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan and Center for Biotechnology, National Taiwan University, Taipei 106, Taiwan,*To whom correspondence should be addressed. Tel: +886 2 23562214; Fax: +886 2 23915295;
| |
Collapse
|
38
|
Wlodkowic D, Skommer J, Akagi J, Fujimura Y, Takeda K. Multiparameter analysis of apoptosis using lab-on-a-chip flow cytometry. CURRENT PROTOCOLS IN CYTOMETRY 2013; 66:9.42.1-9.42.15. [PMID: 24510726 DOI: 10.1002/0471142956.cy0942s66] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The age of microfluidic flow cytometry (µFCM) is fast becoming a reality. One of the most exciting applications of miniaturized chip-based cytometers is multivariate analysis using sampling volumes as small as 10 µl while matching the multiparameter data collection of conventional flow cytometers. We outline several innovative protocols for analyzing caspase-dependent cell death and cell cycle (DNA-content) profile using a fully integrated microfluidic flow cytometry system, Fishman-R. The first protocol describes the use of a new plasma membrane-permeability marker, DRAQ7, and the fluorogenic caspase substrate PhiPhiLux to track caspase activation during programmed cell death. Also outlined is the use of DRAQ7 fluorochrome in conjunction with the mitochondrial membrane potential-sensitive probe TMRM to track dissipation of inner mitochondrial cross-membrane potential. Another protocol adds the ability to measure dissipation of mitochondrial inner membrane potential (using TMRM probe) in relation to the cell cycle profile (using DRAQ5 probe) in living leukemic cells. Finally, we describe the combined use of fluorogenic caspases substrate PhiPhiLux with DRAQ5 probe to measure caspase activation in relation to the cell cycle profile in living tumor cells.
Collapse
Affiliation(s)
- Donald Wlodkowic
- The BioMEMS Research Group, School of Chemical Sciences, University of Auckland, Auckland, New Zealand
- The BioMEMS Research Group, School of Applied Sciences, RMIT University, Melbourne, Australia
| | - Joanna Skommer
- The BioMEMS Research Group, School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Jin Akagi
- The BioMEMS Research Group, School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - Yoo Fujimura
- R&D Division, On-chip Biotechnologies, Tokyo, Japan
| | - Kazuo Takeda
- R&D Division, On-chip Biotechnologies, Tokyo, Japan
| |
Collapse
|
39
|
Awasthi P, Dogra S, Barthwal R. Multispectroscopic methods reveal different modes of interaction of anti cancer drug mitoxantrone with Poly(dG-dC).Poly(dG-dC) and Poly(dA-dT).Poly(dA-dT). JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2013; 127:78-87. [PMID: 23968995 DOI: 10.1016/j.jphotobiol.2013.07.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 07/20/2013] [Accepted: 07/24/2013] [Indexed: 11/29/2022]
Abstract
The interaction of mitoxantrone with alternating Poly(dG-dC).Poly(dG-dC) and Poly(dA-dT).Poly(dA-dT) duplex has been studied by absorption, fluorescence and Circular Dichroism (CD) spectroscopy at Drug to Phosphate base pair ratios D/P=20.0-0.04. Binding to GC polymer occurs in two distinct modes: partial stacking characterized by red shifts of 18-23nm at D/P=0.2-0.8 and external binding at D/P=1.0-20.0 whereas that to AT polymer occurs externally in the entire range of D/P. The binding constant and number of binding sites is 3.7×10(5)M(-1), 0.3 and 1.3× 10(4)M(-1), 1.5 in GC and AT polymers, respectively at low D/P ratios. CD binding isotherms show breakpoints at D/P=0.1, 0.5 and 0.25, 0.5 in GC and AT polymers, respectively. The intrinsic CD bands indicate that the distortions in GC polymer are significantly higher than that in AT polymer. Docking studies show partial insertion of mitoxantrone rings between to GC base pairs in alternating GC polymer. Side chains of mitoxantrone interact specifically with base pairs and DNA backbone. The studies are relevant to the understanding of suppression or inhibition of DNA cleavage on formation of ternary complex with topoisomerase-II enzyme and hence the anti cancer action.
Collapse
Affiliation(s)
- Pamita Awasthi
- Department of Chemistry, National Institute of Technology Hamirpur, Hamirpur 177 001, India
| | | | | |
Collapse
|
40
|
Zhao H, Halicka HD, Li J, Darzynkiewicz Z. Berberine suppresses gero-conversion from cell cycle arrest to senescence. Aging (Albany NY) 2013; 5:623-36. [PMID: 23974852 PMCID: PMC3796215 DOI: 10.18632/aging.100593] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 08/20/2013] [Indexed: 01/05/2023]
Abstract
Berberine (BRB), a natural alkaloid, has a long history of medicinal use in both Ayurvedic and old Chinese medicine. Recently, available as a dietary supplement, Berberine is reported to have application in treatment of variety diseases. Previously we observed that BRB inhibited mTOR/S6 signaling concurrently with reduction of the level of endogenous oxidants and constitutive DNA damage response. We currently tested whether Berberine can affect premature, stress-induced cellular senescence caused by mitoxantrone. The depth of senescence was quantitatively measured by morphometric parameters, senescence-associated β-galactosidase, induction of p21WAF1, replication stress (γH2AX expression), and mTOR signaling; the latter revealed by ribosomal S6 protein (rpS6) phosphorylation. All these markers of senescence were distinctly diminished, in a concentration-dependent manner, by Berberine. In view of the evidence that BRB localizes in mitochondria, inhibits respiratory electron chain and activates AMPK, the observed attenuation of the replication stress-induced cellular senescence most likely is mediated by AMPK that leads to inhibition of mTOR signaling. In support of this mechanism is the observation that rhodamine123, the cationic probe targeting mitochondrial electron chain, also suppressed rpS6 phosphorylation. The present findings reveal that: (a) in cells induced to senescence BRB exhibits gero-suppressive properties by means of mTOR/S6 inhibition; (b) in parallel, BRB reduces the level of constitutive DNA damage response, previously shown to report oxidative DNA damage by endogenous ROS; (c) there appears to a causal linkage between the (a) and (b) activities; (d) the in vitro model of premature stress-induced senescence can be used to assess effectiveness of potential gero-suppressive agents targeting mTOR/S6 and ROS signaling; (e) since most of the reported beneficial effects of BRB are in age-relate diseases, it is likely that gero-suppression is the primary activity of this traditional medicine.
Collapse
Affiliation(s)
- Hong Zhao
- Brander Cancer Research Institute and Department of Pathology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | |
Collapse
|
41
|
Dogra S, Awasthi P, Tripathi S, Pradeep T, Nair MS, Barthwal R. NMR-based structure of anticancer drug mitoxantrone stacked with terminal base pair of DNA hexamer sequence d-(ATCGAT)2. J Biomol Struct Dyn 2013; 32:1164-83. [DOI: 10.1080/07391102.2013.809021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
42
|
Dogra S, Awasthi P, Nair M, Barthwal R. Interaction of anticancer drug mitoxantrone with DNA hexamer sequence d-(CTCGAG)2 by absorption, fluorescence and circular dichroism spectroscopy. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2013; 123:48-54. [PMID: 23624101 DOI: 10.1016/j.jphotobiol.2013.03.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 03/25/2013] [Accepted: 03/28/2013] [Indexed: 10/27/2022]
Abstract
The interaction of mitoxantrone with d-(CTCGAG)2 has been studied by absorption, fluorescence and circular dichroism (CD) spectroscopy. The hypochromism and quenching of fluorescence showed that about four mitoxantrone molecules may be binding externally to DNA hexamer sequence at high drug to nucleic (D/N) acid duplex ratios (28.0-1.1). At lower D/N ratios (1.0-0.2), a red shift in absorption maxima at 610 and 660 nm by 15 and 20 nm, respectively and a red shift in emission maxima by 11 nm accompanied by increase in absorbance and emission has been observed. The equilibrium constant for binding at low (1.0-0.2) and high (28.0-1.1) D/N ratios is 1.8×10(5) M(-1) and 1.38×10(6) M(-1), respectively. The CD spectra show change in intensity of bands accompanied by appearance of induced bands at 325 nm and 650-700 nm. The 251 nm band shows blue shift at D/N ratio of 0.25 and 0.5. The binding isotherms show stoichiometry of 0.25 and 0.5 mitoxantrone molecules binding per duplex. The results suggest stacking of aromatic chromophore of mitoxantrone with terminal base pair of DNA strand forming a sandwiched structure of mitoxantrone between four and two duplex molecules. These investigations are relevant to the formation of ternary complex with topoisomerase enzyme and hence an understanding of anti tumor action of mitoxantrone.
Collapse
Affiliation(s)
- Shilpa Dogra
- Department of Chemistry, National Institute of Technology Hamirpur, Hamirpur 177 001, India
| | | | | | | |
Collapse
|
43
|
Buchelnikov AS, Hernandez Santiago AA, Gonzalez Flores M, Vazquez Ramirez R, Davies DB, Evstigneev MP. General analysis of competitive binding in drug-interceptor-DNA systems. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2012; 41:273-83. [PMID: 22213076 DOI: 10.1007/s00249-011-0783-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 12/09/2011] [Accepted: 12/12/2011] [Indexed: 10/14/2022]
Abstract
A general model of competitive binding in drug-interceptor-DNA systems has been developed in order to quantify both the interceptor and protector mechanisms. The model involves full parameterization of the basic equations governing the mutual competition between drugs binding to DNA and incorporates as partial cases various similar models existing in the literature. The generality of the model results from strict accounting of the statistical effects of the binding of the drug and interceptor with DNA according to the McGhee-von Hippel formalism, and to the strict treatment of hetero-association between the drug and interceptor, which includes formation of all possible types of self- and hetero-complexes in solution. Indirect experimental evidence is provided for the importance of the protector mechanism in drug-caffeine-DNA systems, which is sometimes ignored in the literature because of the small magnitude of the CAF-DNA binding constant.
Collapse
Affiliation(s)
- A S Buchelnikov
- Department of Physics, Sevastopol National Technical University, Universitetskaya str., 33, Sevastopol 99053, Ukraine
| | | | | | | | | | | |
Collapse
|
44
|
Zhao H, Rybak P, Dobrucki J, Traganos F, Darzynkiewicz Z. Relationship of DNA damage signaling to DNA replication following treatment with DNA topoisomerase inhibitors camptothecin/topotecan, mitoxantrone, or etoposide. Cytometry A 2012; 81:45-51. [PMID: 22140093 PMCID: PMC3242513 DOI: 10.1002/cyto.a.21172] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 10/13/2011] [Accepted: 10/28/2011] [Indexed: 01/23/2023]
Abstract
DNA topoisomerase I (Top1) and topoisomerase II (Top2) inhibitors are widely used to treat a variety of cancers. Their mechanism of action involves stabilization of otherwise transient ("cleavable") complexes between Top1 or Top2 and DNA; collisions of DNA replication forks with such stabilized complexes lead to formation of DNA double-strand breaks (DSBs). In this study, using 5-ethynyl-2'deoxyuridine (EdU) as a DNA precursor, we directly assessed the relationship between DNA replication and induction of DSBs revealed as γH2AX foci in A549 cells treated with Top1 inhibitors topotecan (Tpt) or camptothecin (Cpt) and Top2 inhibitors mitoxantrone (Mxt) and etoposide (Etp). Analysis of cells by multiparameter laser scanning cytometry following treatment with Tpt or Cpt revealed that only DNA replicating cells showed induction of γH2AX and a strong correlation between DNA replication and formation of DSBs (r = 0.86). In cells treated with Mxt or Etp, the correlation was weaker (r = 0.52 and 0.64). In addition, both Mtx and Etp caused induction of γH2AX in cells not replicating DNA. Confocal imaging of nuclei of cells treated with Tpt revealed the presence of γH2AX foci predominantly in DNA replicating cells and close association and co-localization of γH2AX foci with DNA replication sites. In cells treated with Mxt or Etp, the γH2AX foci were induced in DNA replicating as well as non-replicating cells but the close association between a large proportion of γH2AX foci and DNA replication sites was also apparent. The data are consistent with the view that collision of DNA replication forks with cleavable Top1-DNA complexes stabilized by Tpt/Cpt is the sole cause of induction of DSBs. Additional mechanisms such as involvement of transcription and/or generation of oxidative stress may contribute to DSBs induction by Mxt and Etp. The confocal analysis of the association between DNA replication sites and the sites of DSBs (γH2AX foci) opens a new approach for mechanistic studies of the involvement of DNA replication in induction of DNA damage.
Collapse
Affiliation(s)
- Hong Zhao
- Brander Cancer Research Institute and Department of Pathology, New York Medical College, Valhalla, New York 10595
| | - Paulina Rybak
- Division of Cell Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jurek Dobrucki
- Division of Cell Biophysics, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Frank Traganos
- Brander Cancer Research Institute and Department of Pathology, New York Medical College, Valhalla, New York 10595
| | - Zbigniew Darzynkiewicz
- Brander Cancer Research Institute and Department of Pathology, New York Medical College, Valhalla, New York 10595
| |
Collapse
|
45
|
Lu Y, Wang GK, Lv J, Zhang GS, Liu QF. Study on the Interaction of an Anthracycline Disaccharide with DNA by Spectroscopic Techniques and Molecular Modeling. J Fluoresc 2010; 21:409-14. [DOI: 10.1007/s10895-010-0729-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2010] [Accepted: 09/27/2010] [Indexed: 04/10/2023]
|
46
|
Wlodkowic D, Skommer J, Darzynkiewicz Z. Cytometry in cell necrobiology revisited. Recent advances and new vistas. Cytometry A 2010; 77:591-606. [PMID: 20235235 PMCID: PMC2975392 DOI: 10.1002/cyto.a.20889] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Over a decade has passed since publication of the last review on "Cytometry in cell necrobiology." During these years we have witnessed many substantial developments in the field of cell necrobiology such as remarkable advancements in cytometric technologies and improvements in analytical biochemistry. The latest innovative platforms such as laser scanning cytometry, multispectral imaging cytometry, spectroscopic cytometry, and microfluidic Lab-on-a-Chip solutions rapidly emerge as highly advantageous tools in cell necrobiology studies. Furthermore, we have recently gained substantial knowledge on alternative cell demise modes such as caspase-independent apoptosis-like programmed cell death (PCD), autophagy, necrosis-like PCD, or mitotic catastrophe, all with profound connotations to pathogenesis and treatment. Although detection of classical, caspase-dependent apoptosis is still the major ground for the advancement of cytometric techniques, there is an increasing demand for novel analytical tools to rapidly quantify noncanonical modes of cell death. This review highlights the key developments warranting a renaissance and evolution of cytometric techniques in the field of cell necrobiology.
Collapse
Affiliation(s)
- Donald Wlodkowic
- The Bioelectronics Research Centre, University of Glasgow, Glasgow, United Kingdom.
| | | | | |
Collapse
|
47
|
Gianoncelli A, Basili S, Scalabrin M, Sosic A, Moro S, Zagotto G, Palumbo M, Gresh N, Gatto B. Rational Design, Synthesis, and DNA Binding Properties of Novel Sequence-Selective Peptidyl Congeners of Ametantrone. ChemMedChem 2010; 5:1080-91. [DOI: 10.1002/cmdc.201000106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
48
|
Coupled spectral and electrochemical evaluation of the anticancer drug mitoxantrone–sodium dodecyl sulfate interaction. Int J Pharm 2010; 390:100-6. [DOI: 10.1016/j.ijpharm.2009.12.064] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 12/10/2009] [Accepted: 12/13/2009] [Indexed: 11/21/2022]
|
49
|
Lu Y, Lv J, Zhang G, Wang G, Liu Q. Interaction of an anthracycline disaccharide with ctDNA: Investigation by spectroscopic technique and modeling studies. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2010; 75:1511-1515. [PMID: 20197239 DOI: 10.1016/j.saa.2010.02.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 01/17/2010] [Accepted: 02/03/2010] [Indexed: 05/28/2023]
Abstract
This study was designed to examine the interaction of an anthracycline disaccharide, 4'-O-(beta-L-oleandrosyl) daunorubicin (DNR-D2), with calf thymus deoxyribonucleic acid (ctDNA) by UV-vis in combination with fluorescence spectroscopy and molecular modeling techniques under physiological conditions (Britton-Robinson buffer solutions, pH 7.4). By the analysis of UV-vis and fluorescence spectrum, it was observed that the binding mode between DNR-D2 and ctDNA might be intercalation, and fluorescence quenching mechanism of DNR-D2 by ctDNA was a static quenching type. Upon binding to ctDNA, the anthraquinone chromophore of DNR-D2 could slide into the C-G rich region of ctDNA. Hydrogen bonding forces may play an essential role in the binding of DNR-D2 to ctDNA. Furthermore, the results obtained from computational modeling corroborated the experimental results obtained from spectroscopic investigations. These studies are valuable for a better understanding the datailed mode of DNR-D2-DNA interaction, which should be important in deeper insight into the therapeutic efficiency of DNR-D2.
Collapse
Affiliation(s)
- Yan Lu
- School of Chemistry and Environmental Science, Henan Normal University, Xinxiang 453007, China.
| | | | | | | | | |
Collapse
|
50
|
Zhao H, Traganos F, Dobrucki J, Wlodkowic D, Darzynkiewicz Z. Induction of DNA damage response by the supravital probes of nucleic acids. Cytometry A 2009; 75:510-9. [PMID: 19373929 DOI: 10.1002/cyto.a.20727] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The aim of this study was to assess the potential DNA damage response (DDR) to four supravitally used biomarkers Hoechst 33342 (Ho 42), DRAQ5, DyeCycle Violet (DCV), and SYTO 17. A549 cells were exposed to these biomarkers at concentrations generally applied to live cells and their effect on histone H2AX (Ser 139), p53 (Ser15), ATM (Ser1981), and Chk2 (Thr68) phosphorylation was assessed using phospho-specific Abs. Short-term treatment with Ho 42 led to modest degree of ATM activation with no evidence of H2AX, Chk2, or p53 phosphorylation. However, pronounced ATM, Chk2, and p53 phosphorylation and perturbed G(2) progression were seen after 18 h. While short-term treatment with DRAQ5 induced ATM activation with no effect on H2AX, Chk2, and p53, dramatic changes marked by a high degree of H2AX, ATM, Chk2, and p53 phosphorylation, all occurring predominantly in S phase cells, and a block in cell cycle progression, were seen after 18 h exposure. These changes suggest that the DRAQ5-induced DNA lesions may become converted into double-strand DNA breaks during replication. Exposure to DCV also led to an increase in the level of activated ATM and Chk2 as well as of phosphorylated p53 and accumulation of cells in G(2)M and S phase. Exposure to SYTO 17 had no significant effect on any of the measured parameters. The data indicate that supravital use of Ho 42, DRAQ5, and DCV induces various degrees of DDR, including activation of ATM, Chk2 and p53, which may have significant consequences on regulatory cell cycle pathways and apoptosis.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Pathology, Brander Cancer Research Institute, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | |
Collapse
|