1
|
Ağan K, Kaya ST, Ağan AF, Ağyar-Yoldaş P, Yoldaş T, İkinci-Keleş A, Çaprazlı T, Arıca E, Kekeçoglu M. Alleviating doxorubicin-induced reproductive toxicity: protective and androgenic effects of drone larvae on sperm morphology and hormonal balance. Toxicol Res 2025; 41:149-165. [PMID: 40013081 PMCID: PMC11850692 DOI: 10.1007/s43188-024-00270-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/30/2024] [Accepted: 12/02/2024] [Indexed: 02/28/2025] Open
Abstract
Male infertility and compromised sperm quality are common side effects of Doxorubicin (DOX), a widely used chemotherapy drug. Its detrimental impact on male reproductive cells underscores the urgent need for effective protective measures. Lyophilized drone larvae (DL) from apitherapy have emerged as a potential solution due to their reported protective properties. By exploring DL's therapeutic potential, this research seeks to address the pressing need for strategies to protect male reproductive health during cancer treatment. The study aims to evaluate the protective effects of lyophilized DL from apitherapy against DOX-induced testicular damage in adult Sprague-Dawley rats. DOX negatively impacts male reproductive cells, leading to infertility and compromised sperm quality. Investigating DL's protective properties is crucial for understanding its therapeutic potential in mitigating such adverse effects. Forty rats were divided into four groups: control, DOX-treated, DL-treated, and DOX + DL-treated. Histopathological assessments, biochemical analyses (TAS, TOS, CAT, SOD, GPX), inflammatory marker measurements (TNF-α, IL-1β, IL-6), and comet assays for DNA damage were conducted on testicular tissue and blood samples. DOX induced histopathological alterations in the testis and epididymis, which DL mitigated. DL increased TAS levels, counteracted DOX-induced decreases in glutathione peroxidase (GPx), total protein, albumin, and increases in total cholesterol. DL also mitigated the rise in Follicle-Stimulating Hormone (FSH) levels caused by DOX, while increasing testosterone levels and lowering Luteinizing Hormone (LH) levels. Inflammatory markers remained unaffected. Tail moment measurements indicated a protective effect against DOX-induced DNA damage in erythrocytes with DL. DL protected sperm morphology, count, and Johnsen's score from DOX-induced reductions, suggesting its potential in mitigating cancer treatment side effects on male reproductive health. The findings suggest that DL, as an apitherapy product, holds significant promise in mitigating DOX's adverse effects on male reproductive systems. However, further investigations into its mechanisms and clinical applications in cancer therapy are warranted, emphasizing the need for continued research to fully understand DL's therapeutic benefits.
Collapse
Affiliation(s)
- Kağan Ağan
- Experimental Animals Application and Research Center, Düzce University, Düzce, Türkiye
| | - Salih Tunç Kaya
- Department of Biology, Faculty of Arts and Science, Düzce University, Düzce, Türkiye
| | - Aydan Fülden Ağan
- Experimental Animals Application and Research Center, Düzce University, Düzce, Türkiye
| | - Pınar Ağyar-Yoldaş
- The Scientific and Technological Research Application and Research Center, Düzce University, Düzce, Türkiye
| | - Taner Yoldaş
- The Scientific and Technological Research Application and Research Center, Düzce University, Düzce, Türkiye
| | - Ayşe İkinci-Keleş
- Department of Histology and Embryology, Faculty of Medicine, Aksaray University, Aksaray, Türkiye
| | - Tuğçe Çaprazlı
- Department of Plant and Animal Production, Düzce Vocational School, Beekeeping Programme, Düzce University, Düzce, Türkiye
- Beekeeping Research, Development and Application Centre, Düzce University, Düzce, Türkiye
| | - Elif Arıca
- Centro Laboratuvarlari, Kagithane, Istanbul, Türkiye
| | - Meral Kekeçoglu
- Department of Biology, Faculty of Arts and Science, Düzce University, Düzce, Türkiye
- Beekeeping Research, Development and Application Centre, Düzce University, Düzce, Türkiye
| |
Collapse
|
2
|
Nayak J, P SV, Sahoo SK, Kumar M, Vashistha VK, Kumar R. Computational insight of antioxidant and doxorubicin combination for effective cancer therapy. J Biomol Struct Dyn 2024; 42:7874-7882. [PMID: 37545163 DOI: 10.1080/07391102.2023.2242507] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 07/23/2023] [Indexed: 08/08/2023]
Abstract
Doxorubicin (DOX) is the most effective antineoplastic agent, destroys cancer cells by interrupting cellular function. However, the serious side effects on the heart limits its utility. To curb these unwanted side effects, nutritionist recommend antioxidants use along with DOX while chemotherapy. But it was not supported by various oncologists as it can alter the toxicity of DOX towards cancer cells. Therefore, here we explored the in silico pharmacokinetics and combination effect of DOX and antioxidants on topoisomerases-II (Top-II) and cyclophilin D (Cyp-D) therapeutic targets involved in cancer proliferation and post-myocardial infarction, respectively. The molecular docking study was conducted on target proteins and DOX including most prescribed antioxidants (melatonin, N-acetylcysteine (NAC), glutathione (GSH), β-carotene and vitamin C). GSH showed effective binding potential for Top-II and Cyp-D active sites, but other considered antioxidants possess low binding affinity. The highest docked conformations were subjected to molecular dynamics (MD) simulations to understand conformer stability of DOX and GSH with Cyp-D and Top-II for 100 ns. The results revealed that ligands pose at Top-II active sites where DOX showed strong binding affinity to DNA binding pocket and GSH to a buried site. The computational data summarised and proposed the GSH and DOX combination as antagonist effects on Top-II. Conversely, the binding compactness of GSH improved due to surface fit at the active pocket of Cyp-D and completely blocking DOX binding affinity, suppress adverse reactions of post-myocardial infarction.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jyotsnamayee Nayak
- Department of Chemistry, S.V. National Institute of Technology, Gujarat, India
| | - Seshu Vardhan P
- Department of Chemistry, S.V. National Institute of Technology, Gujarat, India
| | - Suban K Sahoo
- Department of Chemistry, S.V. National Institute of Technology, Gujarat, India
| | - Manish Kumar
- Department of Chemistry and Chemical Science, School of Physical and Material Sciences, Central University of Himachal Pradesh, Kangra, Himachal Pradesh, India
| | | | - Rajender Kumar
- Department of Chemistry and Chemical Science, School of Physical and Material Sciences, Central University of Himachal Pradesh, Kangra, Himachal Pradesh, India
| |
Collapse
|
3
|
Zhao CL, Mou HZ, Pan JB, Xing L, Mo Y, Kang B, Chen HY, Xu JJ. AI-assisted mass spectrometry imaging with in situ image segmentation for subcellular metabolomics analysis. Chem Sci 2024; 15:4547-4555. [PMID: 38516065 PMCID: PMC10952063 DOI: 10.1039/d4sc00839a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Subcellular metabolomics analysis is crucial for understanding intracellular heterogeneity and accurate drug-cell interactions. Unfortunately, the ultra-small size and complex microenvironment inside the cell pose a great challenge to achieving this goal. To address this challenge, we propose an artificial intelligence-assisted subcellular mass spectrometry imaging (AI-SMSI) strategy with in situ image segmentation. Based on the nanometer-resolution MSI technique, the protonated guanine and threonine ions were respectively employed as the nucleus and cytoplasmic markers to complete image segmentation at the subcellular level, avoiding mutual interference of signals from various compartments in the cell. With advanced AI models, the metabolites within the different regions could be further integrated and profiled. Through this method, we decrypted the distinct action mechanism of isomeric drugs, doxorubicin (DOX) and epirubicin (EPI), only with a stereochemical inversion at C-4'. Within the cytoplasmic region, fifteen specific metabolites were discovered as biomarkers for distinguishing the drug action difference between DOX and EPI. Moreover, we identified that the downregulations of glutamate and aspartate in the malate-aspartate shuttle pathway may contribute to the higher paratoxicity of DOX. Our current AI-SMSI approach has promising applications for subcellular metabolomics analysis and thus opens new opportunities to further explore drug-cell specific interactions for the long-term pursuit of precision medicine.
Collapse
Affiliation(s)
- Cong-Lin Zhao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Han-Zhang Mou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Jian-Bin Pan
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Lei Xing
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Yuxiang Mo
- State Key Laboratory of Low-Dimensional Quantum Physics, Department of Physics, Tsinghua University Beijing 100084 China
| | - Bin Kang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Hong-Yuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| | - Jing-Juan Xu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University Nanjing 210023 China
| |
Collapse
|
4
|
Mejia G, Su L, Pandey P, Jeanne Dit Fouque K, McGoron AJ, Fernandez-Lima F, He J, Mebel AM, Leng F. Anticancer Drug Doxorubicin Spontaneously Reacts with GTP and dGTP. Chem Res Toxicol 2023; 36:660-668. [PMID: 37000908 DOI: 10.1021/acs.chemrestox.2c00367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Here, we reported a spontaneous reaction between anticancer drug doxorubicin and GTP or dGTP. Incubation of doxorubicin with GTP or dGTP at 37 °C or above yields a covalent product: the doxorubicin-GTP or -dGTP conjugate where a covalent bond is formed between the C14 position of doxorubicin and the 2-amino group of guanine. Density functional theory calculations show the feasibility of this spontaneous reaction. Fluorescence imaging studies demonstrate that the doxorubicin-GTP and -dGTP conjugates cannot enter nuclei although they rapidly accumulate in human SK-OV-3 and NCI/ADR-RES cells. Consequently, the doxorubicin-GTP and -dGTP conjugates are less cytotoxic than doxorubicin. We also demonstrate that doxorubicin binds to ATP, GTP, and other nucleotides with a dissociation constant (Kd) in the sub-millimolar range. Since human cells contain millimolar levels of ATP and GTP, these results suggest that doxorubicin may target ATP and GTP, energy molecules that support essential processes in living organisms.
Collapse
Affiliation(s)
- German Mejia
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Linjia Su
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Popular Pandey
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, United States
- Department of Physics, Florida International University, Miami, Florida 33199, United States
| | - Kevin Jeanne Dit Fouque
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Anthony J McGoron
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, United States
- Department of Biomedical Engineering, Florida International University, Miami, Florida 33199, United States
| | - Francisco Fernandez-Lima
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Jin He
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, United States
- Department of Physics, Florida International University, Miami, Florida 33199, United States
| | - Alexander M Mebel
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| | - Fenfei Leng
- Biomolecular Sciences Institute, Florida International University, Miami, Florida 33199, United States
- Department of Chemistry and Biochemistry, Florida International University, Miami, Florida 33199, United States
| |
Collapse
|
5
|
Zhou J, Wang H, Wang W, Ma Z, Chi Z, Liu S. A Cationic Amphiphilic AIE Polymer for Mitochondrial Targeting and Imaging. Pharmaceutics 2022; 15:103. [PMID: 36678732 PMCID: PMC9866158 DOI: 10.3390/pharmaceutics15010103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/29/2022] Open
Abstract
Mitochondria are important organelles that play key roles in generating the energy needed for life and in pathways such as apoptosis. Direct targeting of antitumor drugs, such as doxorubicin (DOX), to mitochondria into cells is an effective approach for cancer therapy and inducing cancer cell death. To achieve targeted and effective delivery of antitumor drugs to tumor cells, to enhance the therapeutic effect, and to reduce the side effects during the treatment, we prepared a cationic amphiphilic polymer with aggregation-induced emission (AIE) characteristic. The polymer could be localized to mitochondria with excellent organelle targeting, and it showed good mitochondrial targeting with low toxicity. The polymer could also self-assemble into doxorubicin-loaded micelles in phosphate buffer, with a particle size of about 4.3 nm, an encapsulation rate of 11.03%, and micelle drug loading that reached 0.49%. The results of in vitro cytotoxicity experiments showed that the optimal dosage was 2.0 μg/mL, which had better inhibitory effect on tumor cells and less biological toxicity on heathy cells. Therefore, the cationic amphiphilic polymer can partially replace expensive commercial mitochondrial targeting reagents, and it can be also used as a drug loading tool to directly target mitochondria in cells for corresponding therapeutic research.
Collapse
Affiliation(s)
| | | | | | | | | | - Siwei Liu
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, Guangzhou 510275, China
| |
Collapse
|
6
|
Xu J, Shamul JG, Kwizera EA, He X. Recent Advancements in Mitochondria-Targeted Nanoparticle Drug Delivery for Cancer Therapy. NANOMATERIALS 2022; 12:nano12050743. [PMID: 35269231 PMCID: PMC8911864 DOI: 10.3390/nano12050743] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023]
Abstract
Mitochondria are critical subcellular organelles that produce most of the adenosine triphosphate (ATP) as the energy source for most eukaryotic cells. Moreover, recent findings show that mitochondria are not only the "powerhouse" inside cells, but also excellent targets for inducing cell death via apoptosis that is mitochondria-centered. For several decades, cancer nanotherapeutics have been designed to specifically target mitochondria with several targeting moieties, and cause mitochondrial dysfunction via photodynamic, photothermal, or/and chemo therapies. These strategies have been shown to augment the killing of cancer cells in a tumor while reducing damage to its surrounding healthy tissues. Furthermore, mitochondria-targeting nanotechnologies have been demonstrated to be highly efficacious compared to non-mitochondria-targeting platforms both in vitro and in vivo for cancer therapies. Moreover, mitochondria-targeting nanotechnologies have been intelligently designed and tailored to the hypoxic and slightly acidic tumor microenvironment for improved cancer therapies. Collectively, mitochondria-targeting may be a promising strategy for the engineering of nanoparticles for drug delivery to combat cancer.
Collapse
Affiliation(s)
- Jiangsheng Xu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (J.X.); (J.G.S.); (E.A.K.)
| | - James G. Shamul
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (J.X.); (J.G.S.); (E.A.K.)
| | - Elyahb Allie Kwizera
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (J.X.); (J.G.S.); (E.A.K.)
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; (J.X.); (J.G.S.); (E.A.K.)
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201, USA
- Correspondence:
| |
Collapse
|
7
|
Deng Y, Ngo DTM, Holien JK, Lees JG, Lim SY. Mitochondrial Dynamin-Related Protein Drp1: a New Player in Cardio-oncology. Curr Oncol Rep 2022; 24:1751-1763. [PMID: 36181612 PMCID: PMC9715477 DOI: 10.1007/s11912-022-01333-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW This study is aimed at reviewing the recent progress in Drp1 inhibition as a novel approach for reducing doxorubicin-induced cardiotoxicity and for improving cancer treatment. RECENT FINDINGS Anthracyclines (e.g. doxorubicin) are one of the most common and effective chemotherapeutic agents to treat a variety of cancers. However, the clinical usage of doxorubicin has been hampered by its severe cardiotoxic side effects leading to heart failure. Mitochondrial dysfunction is one of the major aetiologies of doxorubicin-induced cardiotoxicity. The morphology of mitochondria is highly dynamic, governed by two opposing processes known as fusion and fission, collectively known as mitochondrial dynamics. An imbalance in mitochondrial dynamics is often reported in tumourigenesis which can lead to adaptive and acquired resistance to chemotherapy. Drp1 is a key mitochondrial fission regulator, and emerging evidence has demonstrated that Drp1-mediated mitochondrial fission is upregulated in both cancer cells to their survival advantage and injured heart tissue in the setting of doxorubicin-induced cardiotoxicity. Effective treatment to prevent and mitigate doxorubicin-induced cardiotoxicity is currently not available. Recent advances in cardio-oncology have highlighted that Drp1 inhibition holds great potential as a targeted mitochondrial therapy for doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Yali Deng
- Department of Surgery and Medicine, University of Melbourne, Melbourne, Victoria Australia ,O’Brien Institute Department, St Vincent’s Institute of Medical Research, Fitzroy, Victoria Australia
| | - Doan T. M. Ngo
- School of Biomedical Science and Pharmacy, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, New Lambton Heights, New South Wales Australia
| | - Jessica K. Holien
- Department of Surgery and Medicine, University of Melbourne, Melbourne, Victoria Australia ,School of Science, STEM College, RMIT University, Melbourne, Victoria Australia
| | - Jarmon G. Lees
- Department of Surgery and Medicine, University of Melbourne, Melbourne, Victoria Australia ,O’Brien Institute Department, St Vincent’s Institute of Medical Research, Fitzroy, Victoria Australia
| | - Shiang Y. Lim
- Department of Surgery and Medicine, University of Melbourne, Melbourne, Victoria Australia ,O’Brien Institute Department, St Vincent’s Institute of Medical Research, Fitzroy, Victoria Australia ,Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Victoria Australia ,National Heart Research Institute Singapore, National Heart Centre, Singapore, Singapore
| |
Collapse
|
8
|
Waz S, Matouk AI. Cardioprotective effect of allyl isothiocyanate in a rat model of doxorubicin acute toxicity. Toxicol Mech Methods 2021; 32:194-203. [PMID: 34635025 DOI: 10.1080/15376516.2021.1992064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Doxorubicin (DOX) is an effective anthracycline chemotherapeutic drug. Nevertheless, the cardiotoxicity adverse effect restricts its clinical benefit. Allyl isothiocyanate (AITC) is a natural antioxidant and anti-inflammatory agent. In the present study, we investigated the effect of AITC on cardiotoxicity of DOX. Thirty-two adult male albino rats were divided into four groups; control, AITC, DOX, and AITC + DOX. AITC was administrated orally (25 mg/kg/day) for 7 days, and DOX was given as a single i.p. injection (15 mg/kg) on the third day. Mortality rate was observed during the experiment. Cardiac toxicity markers (lactate dehydrogenase (LDH), creatine kinase (CK-MB), and cardiac Troponin I (cTn-I)) were evaluated in serum samples obtained from all groups after 48 hours of DOX injection. DOX-treated group showed 40% mortality and a significant increase in cardiac enzymes. This increase was accompanied by degenerated cardiomyocytes, and inflammatory cells infiltrates. Interestingly, AITC administration alleviated myocardial oxidative stress induced by DOX as attenuated the increase in malondialdehyde (MDA), and nitric oxide (NO) while resulted in elevations of the antioxidant reduced glutathione (GSH) level as well as superoxide dismutase (SOD) activity. Furthermore, the inflammatory cytokine, TNF-α, was reduced upon administration of AITC with DOX. The cardio-protection of AITC is attributed to increase the expression of cytoprotective nuclear factor erythroid 2-related factor 2 (Nrf2). Subsequently, heme oxygenase 1 (HO-1) level was elevated by AITC to correct the oxidative stress induced by DOX in the heart. Accordingly, AITC ameliorated acute cardiotoxicity associated with DOX treatment via attenuation of oxidative stress and the induced-tissue inflammatory injury. Abbreviations: DOX: doxrubicin; Nrf2: nuclear factor erythroid 2-related factor 2; HO-1: heme oxygenase 1; AITC: ally isothiocyanate; MDA: malondialdehyde; SOD: superoxide dismutase; GSH: reduced glutathione; TNF-α: tumor necrosis factor alpha.
Collapse
Affiliation(s)
- Shaimaa Waz
- Department of Biochemistry, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| | - Asmaa I Matouk
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, El-Minia, Egypt
| |
Collapse
|
9
|
Kiani Nejad Z, Mirzaei-Kalar Z, Khandar AA. Synthesis of ZnFe2O4@SiO2 nanoparticles as a pH-sensitive drug release system and good nano carrier for CT-DNA binding. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.117155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
10
|
Quantum dots as targeted doxorubicin drug delivery nanosystems in human lung cancer cells. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00077-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Abstract
Background
Lung cancer is one of the most frequently diagnosed cancers all over the world and is also one of the leading causes of cancer-related mortality. The main treatment option for small cell lung cancer, conventional chemotherapy, is characterized by a lack of specificity, resulting in severe adverse effects. Therefore, this study aimed at developing a new targeted drug delivery (TDD) system based on Ag–In–Zn–S quantum dots (QDs). For this purpose, the QD nanocrystals were modified with 11-mercaptoundecanoic acid (MUA), L-cysteine, and lipoic acid decorated with folic acid (FA) and used as a novel TDD system for targeting doxorubicin (DOX) to folate receptors (FARs) on adenocarcinomic human alveolar basal epithelial cells (A549). NIH/3T3 cells were used as FAR-negative controls. Comprehensive physicochemical, cytotoxicity, and genotoxicity studies were performed to characterize the developed novel TDDs.
Results
Fourier transformation infrared spectroscopy, dynamic light scattering, and fluorescence quenching confirmed the successful attachment of FA to the QD nanocrystals and of DOX to the QD–FA nanocarriers. UV–Vis analysis helped in determining the amount of FA and DOX covalently anchored to the surface of the QD nanocrystals. Biological screening revealed that the QD–FA–DOX nanoconjugates had higher cytotoxicity in comparison to the other forms of synthesized QD samples, suggesting the cytotoxic effect of DOX liberated from the QD constructs. Contrary to the QD–MUA–FA–DOX nanoconjugates which occurred to be the most cytotoxic against A549 cells among others, no such effect was observed for NIH/3T3 cells, confirming FARs as molecular targets. In vitro scratch assay also revealed significant inhibition of A549 cell migration after treatment with QD–MUA–FA–DOX. The performed studies evidenced that at IC50 all the nanoconjugates induced significantly more DNA breaks than that observed in nontreated cells. Overall, the QD–MUA–FA–DOX nanoconjugates showed the greatest cytotoxicity and genotoxicity, while significantly inhibiting the migratory potential of A549 cells.
Conclusion
QD–MUA–FA–DOX nanoconjugates can thus be considered as a potential drug delivery system for the effective treatment of adenocarcinomic human alveolar basal epithelial cells.
Collapse
|
11
|
Mandić L, Benčić P, Mlinarić-Majerski K, Liekens S, Snoeck R, Andrei G, Kralj M, Basarić N. Substituted adamantylphthalimides: Synthesis, antiviral and antiproliferative activity. Arch Pharm (Weinheim) 2020; 353:e2000024. [PMID: 32285536 DOI: 10.1002/ardp.202000024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 11/07/2022]
Abstract
In this study, three groups of adamantylphthalimides, bearing different substituents at the phthalimide moiety, N-(4'-R2 )phthalimidoadamantanes (1-7), 3-[N-(4'-R2 )phthalimido]-1-adamantanols (8-10), and 3-[N-(4'-R2 )phthalimido]adamantane-1-carboxylic acids (11-15), were synthesized and screened against tumor cells and viruses. The most potent compounds are not substituted at the adamantane and bear an OH or NH2 substituent at the phthalimide (compounds 3 and 5). The antiproliferative activities of compounds 3 and 5 are in the micromolar range, much higher than the one of thalidomide. A minor antiviral activity against cytomegalovirus and varicella-zoster virus was found for compounds 3 and 5, but these compounds lacked selectivity. The results presented are important for the rational design of the next-generation compounds with anticancer and antiviral activities.
Collapse
Affiliation(s)
- Leo Mandić
- Department of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Zagreb, Croatia
| | - Patricia Benčić
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Kata Mlinarić-Majerski
- Department of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Zagreb, Croatia
| | - Sandra Liekens
- Laboratory of Virology and Chemotherapy, Rega Institute, KU Leuven, Leuven, Belgium
| | - Robert Snoeck
- Laboratory of Virology and Chemotherapy, Rega Institute, KU Leuven, Leuven, Belgium
| | - Graciela Andrei
- Laboratory of Virology and Chemotherapy, Rega Institute, KU Leuven, Leuven, Belgium
| | - Marijeta Kralj
- Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia
| | - Nikola Basarić
- Department of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
12
|
Mirzaei-Kalar Z, Yavari A, Jouyban A. Increasing DNA binding affinity of doxorubicin by loading on Fe 3O 4 nanoparticles: A multi-spectroscopic study. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 229:117985. [PMID: 31901801 DOI: 10.1016/j.saa.2019.117985] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/14/2019] [Accepted: 12/20/2019] [Indexed: 06/10/2023]
Abstract
Magnetic Fe3O4 nanoparticles were synthesized successfully by co-precipitation method and characterized using XRD, SEM and EDS analyses. Then doxorubicin (DOX, a known anticancer drug) was loaded onto nanoparticles. In vitro DNA interaction of free DOX and loaded DOX onto Fe3O4 nanoparticles (DOX-Fe3O4) was investigated by DNA-viscosity measurements, UV-visible and fluorescence spectroscopies. The obtained values for binding constant of DOX and DOX-Fe3O4 compounds from UV-visible spectroscopies were 0.04 × 105 and 0.68 × 105 L mol-1, respectively, which confirms DOX-Fe3O4 compound have a stronger interaction with CT-DNA compared to DOX. Considerable changes on viscosity of the compounds recommended that their binding mode with CT-DNA is intercalative binding. Fluorescence intensity of DOX and DOX-Fe3O4 was quenched via static process by regular addition of CT-DNA. Thermodynamic parameters suggest that Van der Waals forces and hydrogen bonding for DOX and electrostatic forces for DOX-Fe3O4 are predominantly responsible for interaction with CT-DNA. Competition fluorescence studies were done by Hoechst 33258 as a well-known groove binder and ethidium bromide (EtBr) as a known intercalator probe. Percentage of displacement for EtBr-DNA complex with DOX and DOX-Fe3O4 was 39% and 61%, and for Hoechst-DNA complex was 9% and 5%, respectively. These results confirmed that both compounds are intercalator binders, although DOX-Fe3O4 with a further 22% displacement is a stronger intercalator binder than DOX. The stronger interaction of DOX-Fe3O4 compared to DOX suggests that the current system can be used as a new and effective way to targeted therapy of anticancer drugs.
Collapse
Affiliation(s)
- Zeinab Mirzaei-Kalar
- Department of Materials Engineering and Nanotechnology, Sabalan University of Advanced Technologies (SUAT), Namin, Iran; Department of Advanced Technologies, University of Mohaghegh Ardabili, Namin, Ardabil, Iran.
| | - Azin Yavari
- Department of Inorganic Chemistry, Faculty of Chemistry, University of Tabriz, Tabriz 51666-14766, Iran
| | - Abolghasem Jouyban
- Pharmaceutical Analysis Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Raza Z, Naureen Z. Melatonin ameliorates the drug induced nephrotoxicity: Molecular insights. Nefrologia 2019; 40:12-25. [PMID: 31735377 DOI: 10.1016/j.nefro.2019.06.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Drug-induced nephrotoxicity is a frequent adverse event that can lead to acute or chronic kidney disease and increase the healthcare expenditure. It has high morbidity and mortality incidence in 40-70% of renal injuries and accounts for 66% cases of renal failure in elderly population. OBJECTIVE Amelioration of drug-induced nephrotoxicity has been long soughed to improve the effectiveness of therapeutic drugs. This study was conducted to review the melatonin potential to prevent the pathogenesis of nephrotoxicity induced by important nephrotoxic drugs. METHODS We analyzed the relevant studies indexed in Pubmed, Medline, Scielo and Web of science to explain the molecular improvements following melatonin co-administration with special attention to oxidative stress, inflammation and apoptosis as key players of drug-induced nephrotoxicity. RESULTS A robust consensus among researchers of these studies suggested that melatonin efficiently eradicate the chain reaction of free radical production and induced the endogenous antioxidant enzymes which attenuate the lipid peroxidation of cellular membranes and subcellular oxidative stress in drug-induced nephrotoxicity. This agreement was further supported by the melatonin role in disintegration of inflammatory process through inhibition of principle pro-inflammatory or apoptotic cytokines such as TNF-α and NF-κB. These studies highlighted that alleviation of drug-induced renal toxicity is a function of melatonin potential to down regulate the cellular inflammatory and oxidative injury process and to stimulate the cellular repair or defensive mechanisms. CONCLUSION The comprehensive nephroprotection and safer profile suggests the melatonin to be a useful adjunct to improve the safety of nephrotoxic drugs.
Collapse
Affiliation(s)
- Zohaib Raza
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan.
| | - Zainab Naureen
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| |
Collapse
|
14
|
Ghanbari K, Roshani M, Goicoechea HC, Jalalvand AR. Developing an elegant and integrated electrochemical-theoretical approach for detection of DNA damage induced by 4-nonylphenol. Heliyon 2019; 5:e02755. [PMID: 31720481 PMCID: PMC6839279 DOI: 10.1016/j.heliyon.2019.e02755] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/03/2019] [Accepted: 10/28/2019] [Indexed: 12/18/2022] Open
Abstract
In this work, a novel biosensor was fabricated for detection of DNA damage induced by 4-nonylphenol (NP) and also determination of NP. To achieve this goal, a glassy carbon electrode (GCE) was modified with chitosan (Chit), gold nanoparticles (Au NPs) and DNA-multiwalled carbon nanotubes (DNA-MWCNTs). Then, the DNA-MWCNTs/Au NPs/Chit/GCE was incubated with methylene blue (MB) to obtain MB-DNA-MWCNTs/Au NPs/Chit/GCE in which MB was used as the redox indicator. The modifications applied to the GCE were characterized by cyclic voltammetry (CV), electrochemical impedance spectroscopy (EIS), scanning electron microscopy (SEM), energy dispersive X-ray spectroscopic (EDS) and theoretical evidence. MB is a derivative of anthraquinone which can intercalate into double helix structure of DNA. By treating MB-DNA-MWCNTs/Au NPs/Chit/GCE with NP, a higher R ct was observed because the insertion of the NP may result in a more negative charge environment on the DNA surface which hinders accessibility of [Fe(CN)6]3-/4- anion to the electrode surface. Change in the EIS response of the biosensor in the presence of NP was used to develop a novel system for monitoring the DNA damage induced by NP. The EIS technique was also used to develop a sensitive electroanalytical method for determination of NP.
Collapse
Affiliation(s)
| | - Mahmoud Roshani
- Department of Chemistry, Ilam University, Ilam, Iran
- Corresponding author.
| | - Hector C. Goicoechea
- Laboratorio de Desarrollo Analítico y Quimiometría (LADAQ), C_atedra de Química Analítica I, Universidad Nacional del Litoral, Ciudad Universitaria, CC 242 (S3000ZAA), Santa Fe, Argentina
| | - Ali R. Jalalvand
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Corresponding author.
| |
Collapse
|
15
|
Grebinyk A, Prylutska S, Chepurna O, Grebinyk S, Prylutskyy Y, Ritter U, Ohulchanskyy TY, Matyshevska O, Dandekar T, Frohme M. Synergy of Chemo- and Photodynamic Therapies with C 60 Fullerene-Doxorubicin Nanocomplex. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E1540. [PMID: 31671590 PMCID: PMC6915635 DOI: 10.3390/nano9111540] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/22/2019] [Accepted: 10/27/2019] [Indexed: 12/17/2022]
Abstract
A nanosized drug complex was explored to improve the efficiency of cancer chemotherapy, complementing it with nanodelivery and photodynamic therapy. For this, nanomolar amounts of a non-covalent nanocomplex of Doxorubicin (Dox) with carbon nanoparticle C60 fullerene (C60) were applied in 1:1 and 2:1 molar ratio, exploiting C60 both as a drug-carrier and as a photosensitizer. The fluorescence microscopy analysis of human leukemic CCRF-CEM cells, in vitro cancer model, treated with nanocomplexes showed Dox's nuclear and C60's extranuclear localization. It gave an opportunity to realize a double hit strategy against cancer cells based on Dox's antiproliferative activity and C60's photoinduced pro-oxidant activity. When cells were treated with 2:1 C60-Dox and irradiated at 405 nm the high cytotoxicity of photo-irradiated C60-Dox enabled a nanomolar concentration of Dox and C60 to efficiently kill cancer cells in vitro. The high pro-oxidant and pro-apoptotic efficiency decreased IC50 16, 9 and 7 × 103-fold, if compared with the action of Dox, non-irradiated nanocomplex, and C60's photodynamic effect, correspondingly. Hereafter, a strong synergy of therapy arising from the combination of C60-mediated Dox delivery and C60 photoexcitation was revealed. Our data indicate that a combination of chemo- and photodynamic therapies with C60-Dox nanoformulation provides a promising synergetic approach for cancer treatment.
Collapse
Affiliation(s)
- Anna Grebinyk
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, 15745 Wildau, Germany.
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany.
| | - Svitlana Prylutska
- Taras Shevchenko National University of Kyiv, Volodymyrska 64, 01601 Kyiv, Ukraine.
| | - Oksana Chepurna
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Sergii Grebinyk
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, 15745 Wildau, Germany.
| | - Yuriy Prylutskyy
- Taras Shevchenko National University of Kyiv, Volodymyrska 64, 01601 Kyiv, Ukraine.
| | - Uwe Ritter
- Institute of Chemistry and Biotechnology, University of Technology Ilmenau, Weimarer Straße 25 (Curiebau), 98693 Ilmenau, Germany.
| | - Tymish Y Ohulchanskyy
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Olga Matyshevska
- Palladin Institute of Biochemistry, NAS of Ukraine, Leontovicha Str. 9, 01030 Kyiv, Ukraine.
| | - Thomas Dandekar
- Department of Bioinformatics, Biocenter, University of Würzburg, Am Hubland, 97074 Würzburg, Germany.
| | - Marcus Frohme
- Division Molecular Biotechnology and Functional Genomics, Technical University of Applied Sciences Wildau, Hochschulring 1, 15745 Wildau, Germany.
| |
Collapse
|
16
|
Smuder AJ. Exercise stimulates beneficial adaptations to diminish doxorubicin-induced cellular toxicity. Am J Physiol Regul Integr Comp Physiol 2019; 317:R662-R672. [PMID: 31461307 DOI: 10.1152/ajpregu.00161.2019] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Doxorubicin (DOX) is a highly effective antitumor agent used for the treatment of a wide range of cancers. Unfortunately, DOX treatment results in cytotoxic side effects due to its accumulation within off-target tissues. DOX-induced cellular toxicity occurs as a result of increased oxidative damage, resulting in apoptosis and cell death. While there is no standard-of-care practice to prevent DOX-induced toxicity to healthy organs, exercise has been shown to prevent cellular dysfunction when combined with DOX chemotherapy. Endurance exercise stimulates numerous biochemical adaptations that promote a healthy phenotype in several vulnerable tissues without affecting the antineoplastic properties of DOX. Therefore, for the development of an effective strategy to combat the pathological effects of DOX, it is important to determine the appropriate exercise regimen to prescribe to cancer patients receiving DOX therapy and to understand the mechanisms responsible for exercise-induced protection against DOX toxicity to noncancer cells. This review summarizes the cytotoxic effects of DOX on the heart, skeletal muscle, liver, and kidneys and discusses the current understanding of the clinical benefits of regular physical activity and the potential mechanisms mediating the positive effects of exercise on each organ system.
Collapse
Affiliation(s)
- Ashley J Smuder
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida
| |
Collapse
|
17
|
Chen J, Chen X, Chen X, Sun H, Yang D. SM‑164 enhances the antitumor activity of adriamycin in human U2‑OS cells via downregulation of X‑linked inhibitor of apoptosis protein. Mol Med Rep 2019; 19:5079-5086. [PMID: 31059038 PMCID: PMC6522877 DOI: 10.3892/mmr.2019.10181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 02/28/2019] [Indexed: 11/05/2022] Open
Abstract
The antitumor effects of SM‑164 and adriamycin (ADM) on human osteosarcoma U2‑OS cells, the underlying mechanism are yet to be investigated. In the present study, U2‑OS cells were divided into control, ADM, SM‑164, and ADM + SM‑164 groups. In addition, cells treated with both SM‑164 and ADM were further divided into three subgroups: SM‑164 + ADM, SM‑164 + ADM + vector and SM‑164 + ADM + X‑linked inhibitor of apoptosis protein (XIAP) silencing groups. XIAP expression was achieved via transfection with shRNA lentiviral vectors. Reverse transcription‑quantitative polymerase chain reaction and western blotting were used to detect the expression of caspases‑7, ‑9, and ‑3, poly ADP‑ribose polymerase (PARP), XIAP, cellular inhibitor of apoptosis protein‑1 (cIAP‑1) and survivin. Cell viability and apoptosis were evaluated using MTT and flow cytometry assays, respectively. Compared with the control group, cell viability decreased, while apoptosis was increased in the ADM and SM‑164‑treatment group. ADM and SM‑164 treatment promoted the expression of caspases‑7, ‑9 and ‑3, and PARP, but reduced the expression of XIAP, survivin and cIAP‑1. Compared with ADM + SM‑164 group, XIAP silencing with ADM + SM‑164 treatment further reduced cell viability, promoted apoptosis, increased caspase‑7, ‑9 and ‑3, and PARP expression; however the expression of survivin and cIAP‑1 were reduced. Combined ADM and SM‑164 treatment may be considered as potential therapeutic agent in the treatment of osteosarcoma, possibly via reductions XIAP expression.
Collapse
Affiliation(s)
- Jiangwei Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xuanyin Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaodong Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Haiying Sun
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangxi 210009, P.R. China
| | - Dong Yang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
18
|
Delivering Combination Chemotherapies and Targeting Oncogenic Pathways via Polymeric Drug Delivery Systems. Polymers (Basel) 2019; 11:polym11040630. [PMID: 30959799 PMCID: PMC6523645 DOI: 10.3390/polym11040630] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/22/2019] [Accepted: 03/24/2019] [Indexed: 12/24/2022] Open
Abstract
The side-effects associated with chemotherapy necessitates better delivery of chemotherapeutics to the tumor. Nanoparticles can load higher amounts of drug and improve delivery to tumors, increasing the efficacy of treatment. Polymeric nanoparticles, in particular, have been used extensively for chemotherapeutic delivery. This review describes the efforts made to deliver combination chemotherapies and inhibit oncogenic pathways using polymeric drug delivery systems. Combinations of chemotherapeutics with other drugs or small interfering RNA (siRNA) combinations have been summarized. Special attention is given to the delivery of drug combinations that involve either paclitaxel or doxorubicin, two popular chemotherapeutics in clinic. Attempts to inhibit specific pathways for oncotherapy have also been described. These include inhibition of oncogenic pathways (including those involving HER2, EGFR, MAPK, PI3K/Akt, STAT3, and HIF-1α), augmentation of apoptosis by inhibiting anti-apoptosis proteins (Bcl-2, Bcl-xL, and survivin), and targeting dysregulated pathways such as Wnt/β-catenin and Hedgehog.
Collapse
|
19
|
Xi J, Li M, Jing B, An M, Yu C, Pinnock CB, Zhu Y, Lam MT, Liu H. Long-Circulating Amphiphilic Doxorubicin for Tumor Mitochondria-Specific Targeting. ACS APPLIED MATERIALS & INTERFACES 2018; 10:43482-43492. [PMID: 30479120 PMCID: PMC6893847 DOI: 10.1021/acsami.8b17399] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The mitochondria have emerged as a novel target for cancer chemotherapy primarily due to their central roles in energy metabolism and apoptosis regulation. Here, we report a new molecular approach to achieve high levels of tumor- and mitochondria-selective deliveries of the anticancer drug doxorubicin. This is achieved by molecular engineering, which functionalizes doxorubicin with a hydrophobic lipid tail conjugated by a solubility-promoting poly(ethylene glycol) polymer (amphiphilic doxorubicin or amph-DOX). In vivo, the amphiphile conjugated to doxorubicin exhibits a dual function: (i) it binds avidly to serum albumin and hijacks albumin's circulating and transporting pathways, resulting in prolonged circulation in blood, increased accumulation in tumor, and reduced exposure to the heart; (ii) it also redirects doxorubicin to mitochondria by altering the drug molecule's intracellular sorting and transportation routes. Efficient mitochondrial targeting with amph-DOX causes a significant increase of reactive oxygen species levels in tumor cells, resulting in markedly improved antitumor efficacy than the unmodified doxorubicin. Amphiphilic modification provides a simple strategy to simultaneously increase the efficacy and safety of doxorubicin in cancer chemotherapy.
Collapse
Affiliation(s)
- Jingchao Xi
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, United States
| | - Meng Li
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, United States
| | - Benxin Jing
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, United States
| | - Myunggi An
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, United States
| | - Chunsong Yu
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, United States
| | - Cameron B. Pinnock
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan 48202, United States
| | - Yingxi Zhu
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, United States
| | - Mai T. Lam
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan 48202, United States
| | - Haipeng Liu
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, Michigan 48202, United States
- Department of Oncology, Wayne State University, Detroit, Michigan 48201, United States
- Tumor Biology and Microenvironment Program, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan 48201, United States
| |
Collapse
|
20
|
Zangeneh MM, Norouzi H, Mahmoudi M, Goicoechea HC, Jalalvand AR. Fabrication of a novel impedimetric biosensor for label free detection of DNA damage induced by doxorubicin. Int J Biol Macromol 2018; 124:963-971. [PMID: 30508544 DOI: 10.1016/j.ijbiomac.2018.11.278] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/17/2018] [Accepted: 11/30/2018] [Indexed: 12/11/2022]
Abstract
In this work, a novel impedimetric biosensor has been fabricated for detection of DNA damage induced by doxorubicin (DX). Cytochrome P450 reductase (CPR) is required for electron transfer from nicotinamide adenine dinucleotide phosphate (NADPH) to cytochrome P450 (CP450) which causes DX to undergo a one-electron reduction of the p-quinone residue to form the semiquinone radical resulting in the generation of free hydroxyl radical which causes DNA damage. After modification of bare glassy carbon electrode (GCE) with multiwalled carbon nanotubes (MWCNTs) and chitosan (Ch), CPR and CP450 were co-immobilized onto the surface of Ch/MWCNTs/GCE by cross-linking CPR, CP450 and Ch through addition of glutaraldehyde. Then, the DNA was assembled onto the surface of CPRCP450/Ch/MWCNTs/GCE to fabricate the biosensor (DNA/CPRCP450/Ch/MWCNTs/GCE). Modifications applied to the bare GCE to fabricate the biosensor were characterized by CV, EIS and SEM. The DNA/CPRCP450/Ch/MWCNTs/GCE was treated in the damaging solution (DX + NADPH) which caused a significant DNA damage and the exposed DNA bases reduced the electrostatic repulsion of the negatively charged redox probe leading to Faradaic impedance changes. Performance of the biosensor for detection of DNA damage in the presence of Spinach extract was also examined and finally, an indirect impedimetric method was developed for determination of DX.
Collapse
Affiliation(s)
- Mohammad Mahdi Zangeneh
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran; Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hasan Norouzi
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Majid Mahmoudi
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hector C Goicoechea
- Laboratorio de Desarrollo Analítico y Quimiometría (LADAQ), Cátedra de Química Analítica I, Universidad Nacional del Litoral, Ciudad Universitaria, CC 242, S3000ZAA Santa Fe, Argentina
| | - Ali R Jalalvand
- Research Center of Oils and Fats, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
21
|
Tan P, Lau B, Krishnasamy G, Ng M, Husin L, Ruslan N, Song D, Velaithan V, Okuda K, Patel V. Zebrafish embryonic development-interfering macrolides from Streptomyces californicus impact growth and mitochondrial function in human colorectal cancer cells. Process Biochem 2018. [DOI: 10.1016/j.procbio.2018.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
22
|
Battogtokh G, Cho YY, Lee JY, Lee HS, Kang HC. Mitochondrial-Targeting Anticancer Agent Conjugates and Nanocarrier Systems for Cancer Treatment. Front Pharmacol 2018; 9:922. [PMID: 30174604 PMCID: PMC6107715 DOI: 10.3389/fphar.2018.00922] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022] Open
Abstract
The mitochondrion is an important intracellular organelle for drug targeting due to its key roles and functions in cellular proliferation and death. In the last few decades, several studies have revealed mitochondrial functions, attracting the focus of many researchers to work in this field over nuclear targeting. Mitochondrial targeting was initiated in 1995 with a triphenylphosphonium-thiobutyl conjugate as an antioxidant agent. The major driving force for mitochondrial targeting in cancer cells is the higher mitochondrial membrane potential compared with that of the cytosol, which allows some molecules to selectively target mitochondria. In this review, we discuss mitochondria-targeting ligand-conjugated anticancer agents and their in vitro and in vivo behaviors. In addition, we describe a mitochondria-targeting nanocarrier system for anticancer drug delivery. As previously reported, several agents have been known to have mitochondrial targeting potential; however, they are not sufficient for direct application for cancer therapy. Thus, many studies have focused on direct conjugation of targeting ligands to therapeutic agents to improve their efficacy. There are many variables for optimal mitochondria-targeted agent development, such as choosing a correct targeting ligand and linker. However, using the nanocarrier system could solve some issues related to solubility and selectivity. Thus, this review focuses on mitochondria-targeting drug conjugates and mitochondria-targeted nanocarrier systems for anticancer agent delivery.
Collapse
Affiliation(s)
| | | | | | | | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon, South Korea
| |
Collapse
|
23
|
Mathivathanan L, Yang G, Leng F, Raptis RG. Crystal structure and conformational analysis of doxorubicin nitrate. Acta Crystallogr E Crystallogr Commun 2018; 74:400-405. [PMID: 29765732 PMCID: PMC5947812 DOI: 10.1107/s2056989018002955] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/19/2018] [Indexed: 11/13/2023]
Abstract
Crystal structure determination of doxorubicin nitrate, (DoxH)NO3, systematic name (7S,9S)-7-{[(2R,4S,5S,6S)-4-azaniumyl-5-hy-droxy-6-methyl-oxan-2-yl]-oxy}-6,9,11-trihy-droxy-9-(2-hy-droxy-acet-yl)-4-meth-oxy-8,10-di-hydro-7H-tetra-cen-5,12-dione nitrate, shows two formula units present in the asymmetric unit. In the crystal lattice, hydrogen-bonded pairs of (DoxH+) cations and segregation of the aglycone and sugar moieties are observed. Inspection of mol-ecular overlays reveals that the conformation of (DoxH)NO3 resembles that of DNA-inter-calated, but not of protein-docked (DoxH)+. The structure was refined as a two-component twin.
Collapse
Affiliation(s)
- Logesh Mathivathanan
- Department of Chemistry and Biochemistry and Biomolecular Sciences Institute, Florida International University, 11200 SW 8th St, Miami, FL 33199, USA
| | - Guang Yang
- Department of Chemistry and Biochemistry and Biomolecular Sciences Institute, Florida International University, 11200 SW 8th St, Miami, FL 33199, USA
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, Henan, 450001, People’s Republic of China
| | - Fenfei Leng
- Department of Chemistry and Biochemistry and Biomolecular Sciences Institute, Florida International University, 11200 SW 8th St, Miami, FL 33199, USA
| | - Raphael G. Raptis
- Department of Chemistry and Biochemistry and Biomolecular Sciences Institute, Florida International University, 11200 SW 8th St, Miami, FL 33199, USA
| |
Collapse
|
24
|
Morton AB, Mor Huertas A, Hinkley JM, Ichinoseki-Sekine N, Christou DD, Smuder AJ. Mitochondrial accumulation of doxorubicin in cardiac and diaphragm muscle following exercise preconditioning. Mitochondrion 2018; 45:52-62. [PMID: 29474837 DOI: 10.1016/j.mito.2018.02.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 11/22/2017] [Accepted: 02/15/2018] [Indexed: 12/22/2022]
Abstract
Doxorubicin (DOX) is a highly effective anthracycline antibiotic. Unfortunately, the clinical use of DOX is limited by the risk of deleterious effects to cardiac and respiratory (i.e. diaphragm) muscle, resulting from mitochondrial reactive oxygen species (ROS) production. In this regard, exercise is demonstrated to protect against DOX-induced myotoxicity and prevent mitochondrial dysfunction. However, the protective mechanisms are currently unclear. We hypothesized that exercise may induce protection by increasing the expression of mitochondria-specific ATP-binding cassette (ABC) transporters and reducing mitochondrial DOX accumulation. Our results confirm this finding and demonstrate that two weeks of exercise preconditioning is sufficient to prevent cardiorespiratory dysfunction.
Collapse
Affiliation(s)
- Aaron B Morton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - Andres Mor Huertas
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - J Matthew Hinkley
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | | | - Demetra D Christou
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - Ashley J Smuder
- Department of Exercise Science, University of South Carolina, Columbia, SC, United States.
| |
Collapse
|
25
|
Ak G, Yilmaz H, Güneş A, Hamarat Sanlier S. In vitro and in vivo evaluation of folate receptor-targeted a novel magnetic drug delivery system for ovarian cancer therapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:926-937. [DOI: 10.1080/21691401.2018.1439838] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Güliz Ak
- Department of Biochemistry, Ege University Faculty of Science, Izmir, Turkey
| | - Habibe Yilmaz
- Department of Biochemistry, Ege University Faculty of Science, Izmir, Turkey
| | - Aybike Güneş
- Department of Biochemistry, Ege University Faculty of Science, Izmir, Turkey
| | | |
Collapse
|
26
|
Mohajeri M, Sahebkar A. Protective effects of curcumin against doxorubicin-induced toxicity and resistance: A review. Crit Rev Oncol Hematol 2018; 122:30-51. [PMID: 29458788 DOI: 10.1016/j.critrevonc.2017.12.005] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/28/2017] [Accepted: 12/11/2017] [Indexed: 02/08/2023] Open
Abstract
Doxorubicin (DOX)-induced toxicity and resistance are major obstacles in chemotherapeutic approaches. Despite effective in the treatment of numerous malignancies, some clinicians have voiced concern that DOX has the potential to cause debilitating consequences in organ tissues, especially the heart. The mechanisms of toxicity and resistance are respectively related to induction of reactive oxygen species (ROS) and up-regulation of ATP-binding cassette (ABC) transporter. Curcumin (CUR) with several biological and pharmacological properties is expected to restore DOX-mediated impairments to tissues. This review is intended to address the current knowledge on DOX adverse effects and CUR protective actions in the heart, kidneys, liver, brain, and reproductive organs. Coadministration of CUR and DOX is capable of ameliorating DOX toxicity pertained to antioxidant, apoptosis, autophagy, and mitochondrial permeability.
Collapse
Affiliation(s)
- Mohammad Mohajeri
- Department of Medical Biotechnology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Shivakumar A, Yogendra Kumar MS. Critical Review on the Analytical Mechanistic Steps in the Evaluation of Antioxidant Activity. Crit Rev Anal Chem 2018; 48:214-236. [DOI: 10.1080/10408347.2017.1400423] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
| | - M. S. Yogendra Kumar
- Defence Bio-Engineering and Electromedical Laboratory, Defense Research and Development Organization, Bengaluru, Karnataka, India
| |
Collapse
|
28
|
Li S, Jiang X, Zheng R, Zuo S, Zhao L, Fan G, Fan J, Liao Y, Yu X, Cheng H. An azobenzene-based heteromeric prodrug for hypoxia-activated chemotherapy by regulating subcellular localization. Chem Commun (Camb) 2018; 54:7983-7986. [PMID: 29963672 DOI: 10.1039/c8cc03430c] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An azobenzene-based heteromeric prodrug (hNDP) was prepared for tumor targeted chemotherapy by regulating subcellular localization.
Collapse
|
29
|
Aung LH, Li R, Prabhakar BS, Maker AV, Li P. Mitochondrial protein 18 (MTP18) plays a pro-apoptotic role in chemotherapy-induced gastric cancer cell apoptosis. Oncotarget 2017; 8:56582-56597. [PMID: 28915614 PMCID: PMC5593585 DOI: 10.18632/oncotarget.17508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 03/27/2017] [Indexed: 02/06/2023] Open
Abstract
One of the severe limitations of chemotherapy is the development of drug resistance. However, the mechanisms underlying chemotherapy resistance remain to be elucidated. Mitochondrial mediated apoptosis is a form of cell death induced by chemotherapy. Several chemotherapeutic agents have been shown to induce mitochondrial fission, and finally activate the apoptosis cascade in various cancer cells. Here, we report that the mitochondrial membrane protein 18 (MTP18) induced mitochondrial fragmentation in gastric cancer cells under doxorubicin (DOX) exposure. Upon over-expression of MTP18, a sub-cytotoxic dose of DOX could sensitize a significant number of cells to undergo mitochondrial fission and subsequent apoptosis. These findings suggest that MTP18 can enhance the sensitivity of gastric cancer cells to DOX. Mechanistically, we found that MTP18 enriched dynamic-related protein 1 (DRP1) accumulation in mitochondria and it was responsible for mediating DOX-induced signaling required for mitochondrial fission. Intriguingly, MTP18 expression was downregulated during DOX treatment. Thus, down-regulation of MTP18 expression could be one of the resistance factors interfering with DOX-induced apoptosis in gastric cancer cells.
Collapse
Affiliation(s)
- Lynn H.H. Aung
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ruibei Li
- School of Professional Studies, Northwestern University, Chicago, IL, USA
| | - Bellur S. Prabhakar
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Ajay V. Maker
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
- Division of Surgical Oncology, Department of Surgery, University of Illinois at Chicago, Creticos Cancer Center, Advocate Illinois Masonic Medical Center, Chicago, IL, USA
| | - Peifeng Li
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
30
|
Cui H, Huan ML, Ye WL, Liu DZ, Teng ZH, Mei QB, Zhou SY. Mitochondria and Nucleus Dual Delivery System To Overcome DOX Resistance. Mol Pharm 2017; 14:746-756. [DOI: 10.1021/acs.molpharmaceut.6b01016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Han Cui
- Department
of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032, China
| | - Meng-lei Huan
- Department
of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032, China
| | - Wei-liang Ye
- Department
of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032, China
| | - Dao-zhou Liu
- Department
of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032, China
| | - Zeng-hui Teng
- Department
of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032, China
| | - Qi-Bing Mei
- Key
Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica
of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi’an, 710032, China
| | - Si-yuan Zhou
- Department
of Pharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi’an, 710032, China
- Key
Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica
of the State Administration of Traditional Chinese Medicine, Fourth Military Medical University, Xi’an, 710032, China
| |
Collapse
|
31
|
Mai Y, Yu JJ, Bartholdy B, Xu-Monette ZY, Knapp EE, Yuan F, Chen H, Ding BB, Yao Z, Das B, Zou Y, Young KH, Parekh S, Ye BH. An oxidative stress-based mechanism of doxorubicin cytotoxicity suggests new therapeutic strategies in ABC-DLBCL. Blood 2016; 128:2797-2807. [PMID: 27737889 PMCID: PMC5159702 DOI: 10.1182/blood-2016-03-705814] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 10/03/2016] [Indexed: 12/16/2022] Open
Abstract
Diffuse large B-cell lymphomas (DLBCLs) contain 2 major molecular subtypes; namely, the germinal center B-cell-like (GCB) and the activated B-cell-like (ABC) DLBCLs. It is well documented that ABC-DLBCL cases have a significantly poorer survival response than GCB-DLBCLs in both the CHOP (cyclophosphamide, vincristine, doxorubicin, and prednisone) and the rituximab (R)-CHOP eras. However, the underlying cause of this subtype disparity is poorly understood. Nevertheless, these clinical observations raise the possibility for an ABC-DLBCL-specific resistance mechanism that is directed toward 1 of the CHOP components and is inadequately addressed by rituximab. Here, we report that the main cytotoxic ingredient in CHOP, doxorubicin (Dox), has subtype-specific mechanisms of cytotoxicity in DLBCLs resulting from differences in the subcellular distribution pattern. Specifically, in cell line models of ABC-DLBCL, Dox is often enriched in the cytoplasm away from the nuclear DNA. As a result, Dox-induced cytotoxicity in ABC-DLBCLs is often dependent on oxidative stress, rather than DNA damage response. These findings are corroborated by gene signature analysis, which demonstrates that basal oxidative stress status predicts treatment outcome among patients with ABC-DLBCL, but not patients with GCB-DLBCL. In terms of redox-related resistance mechanism, our results suggest that STAT3 confers Dox resistance in ABC-DLBCLs by reinforcing an antioxidant program featuring upregulation of the SOD2 gene. Furthermore, a small-molecule STAT3 inhibitor synergizes with CHOP to trigger oxidative stress and kill ABC-DLBCL cells in preclinical models. These results provide a mechanistic basis for development of novel therapies that target either STAT3 or redox homeostasis to improve treatment outcomes for ABC-DLBCLs.
Collapse
Affiliation(s)
- Yun Mai
- Department of Cell Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
| | - J Jessica Yu
- Department of Cell Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
| | - Boris Bartholdy
- Department of Cell Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
| | - Zijun Y Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Esther E Knapp
- Department of Pediatrics
- The Children's Hospital at Montefiore
| | - Fei Yuan
- Department of Cell Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
| | - Hongshan Chen
- Department of Cell Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
| | - B Belinda Ding
- Department of Cell Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
| | - Zhihua Yao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Yiyu Zou
- Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
| | - Ken He Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Samir Parekh
- Department of Cell Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
- Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
| | - B Hilda Ye
- Department of Cell Biology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY
| |
Collapse
|
32
|
Kavazis AN, Morton AB, Hall SE, Smuder AJ. Effects of doxorubicin on cardiac muscle subsarcolemmal and intermyofibrillar mitochondria. Mitochondrion 2016; 34:9-19. [PMID: 27832997 DOI: 10.1016/j.mito.2016.10.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/03/2016] [Accepted: 10/31/2016] [Indexed: 12/27/2022]
Abstract
Doxorubicin (DOX) is a highly effective chemotherapeutic used in the treatment of a broad spectrum of malignancies. However, clinical use of DOX is highly limited by cumulative and irreversible cardiomyopathy that occurs following DOX treatment. The pathogenesis of DOX-induced cardiac muscle dysfunction is complex. However, it has been proposed that the etiology of this myopathy is related to mitochondrial dysfunction, as a result of the dose-dependent increase in the mitochondrial accumulation of DOX. In this regard, cardiac muscle possesses two morphologically distinct populations of mitochondria. Subsarcolemmal (SS) mitochondria are localized just below the sarcolemma, whereas intermyofibrillar (IMF) mitochondria are found between myofibrils. Mitochondria in both regions exhibit subtle differences in biochemical properties, giving rise to differences in respiration, lipid composition, enzyme activities and protein synthesis rates. Based on the heterogeneity of SS and IMF mitochondria, we hypothesized that acute DOX administration would have distinct effects on each cardiac mitochondrial subfraction. Therefore, we isolated SS and IMF mitochondria from the hearts of female Sprague-Dawley rats 48h after administration of DOX. Our results demonstrate that while SS mitochondria appear to accumulate greater amounts of DOX, IMF mitochondria demonstrate a greater apoptotic and autophagic response to DOX exposure. Thus, the divergent protein composition and function of the SS and IMF cardiac mitochondria result in differential responses to DOX, with IMF mitochondria appearing more susceptible to damage after DOX treatment.
Collapse
Affiliation(s)
| | - Aaron B Morton
- Department of Applied Physiology and Kinesiology, University of Florida, Room 25 Florida Gym, , Gainesville, FL 32611, United States
| | - Stephanie E Hall
- Department of Kinesiology, Boise State University, Boise, ID, United States
| | - Ashley J Smuder
- Department of Applied Physiology and Kinesiology, University of Florida, Room 25 Florida Gym, , Gainesville, FL 32611, United States.
| |
Collapse
|
33
|
Mielańczyk A, Skonieczna M, Mielańczyk Ł, Neugebauer D. In Vitro Evaluation of Doxorubicin Conjugates Based on Sugar Core Nonlinear Polymethacrylates toward Anticancer Drug Delivery. Bioconjug Chem 2016; 27:893-904. [PMID: 26942938 DOI: 10.1021/acs.bioconjchem.5b00671] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
V-shaped and star-shaped hydroxylamine-functionalized polymethacrylates designed as nanosized conjugates (<120 nm) with anticancer agent, namely, doxorubicin (DOX), were evaluated in vitro toward their potential usage as drug delivery systems in breast cancer (MCF-7) treatment. Statistical analysis of MTS assay results showed that the 4-arm conjugate (n(DOX) = 16) was the most effective polymeric system against MCF-7/W (wild type) and MCF-7/R (DOX resistant) cell lines. Apoptosis assay analysis showed that MCF-7/R cells cultured with nonlinear copolymers died due to necrosis and late apoptotis, whereas MCF-7/W cells were in early and late apoptosis. Among all tested conjugates, the most promising results with induction of apoptosis without inducing necrosis in both MCF-7 cell lines were obtained for conjugate based on 4-arm stars with low content of DOX. The cell cycle assay revealed that increase of MMA units in 4-arm copolymers induced MCF-7/R cell arrest in the SubG1 phase. In the same cell line, the corresponding conjugates triggered S and G2/M arrest. Gradual internalization of the chosen conjugate by MCF-7/R cells was monitored via fluorescence microscopy showing its main localization in the cytoplasm.
Collapse
Affiliation(s)
- A Mielańczyk
- Department of Physical Chemistry and Technology of Polymers, Faculty of Chemistry, Silesian University of Technology , M. Strzody 9 Street, 44-100 Gliwice, Poland
| | - M Skonieczna
- Biosystems Group, Institute of Automatic Control, Silesian University of Technology , Akademicka 16 Street, 44-100 Gliwice, Poland
| | - Ł Mielańczyk
- School of Medicine with the Division of Dentistry in Zabrze, Department of Histology and Embryology, Medical University of Silesia , Jordana 19 Street, 41-808 Zabrze, Poland
| | - D Neugebauer
- Department of Physical Chemistry and Technology of Polymers, Faculty of Chemistry, Silesian University of Technology , M. Strzody 9 Street, 44-100 Gliwice, Poland
| |
Collapse
|
34
|
Rharass T, Gbankoto A, Canal C, Kurşunluoğlu G, Bijoux A, Panáková D, Ribou AC. Oxidative stress does not play a primary role in the toxicity induced with clinical doses of doxorubicin in myocardial H9c2 cells. Mol Cell Biochem 2016; 413:199-215. [PMID: 26833193 DOI: 10.1007/s11010-016-2653-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 01/23/2016] [Indexed: 11/26/2022]
Abstract
The implication of oxidative stress as primary mechanism inducing doxorubicin (DOX) cardiotoxicity is still questionable as many in vitro studies implied supra-clinical drug doses or unreliable methodologies for reactive oxygen species (ROS) detection. The aim of this study was to clarify whether oxidative stress is involved in compliance with the conditions of clinical use of DOX, and using reliable tools for ROS detection. We examined the cytotoxic mechanisms of 2 μM DOX 1 day after the beginning of the treatment in differentiated H9c2 rat embryonic cardiac cells. Cells were exposed for 2 or 24 h with DOX to mimic a single chronic dosage or to favor accumulation, respectively. We found that apoptosis was prevalent in cells exposed for a short period with DOX: cells showed typical hallmarks as loss of anchorage ability, mitochondrial hyperpolarization followed by the collapse of mitochondrial activity, and nuclear condensation. Increasing the exposure period favored a shift to necrosis as the cells preferentially exhibited early DNA impairment and nuclear swelling. In either case, measuring the fluorescence lifetime of 1-pyrenebutyric acid or the intensities of dihydroethidium or amplex red showed a consistent pattern in ROS production which was a slight increased level far from representative of an oxidative stress. Moreover, pre-treatment with dexrazoxane provided a cytoprotective effect although it failed to detoxify ROS. Our data support that oxidative stress is unlikely to be the primary mechanism of DOX cardiac toxicity in vitro.
Collapse
Affiliation(s)
- Tareck Rharass
- Institute of Modeling and Analysis in Geo-Environmental and Health (IMAGES_ESPACE-DEV), University of Perpignan Via Domitia, 66860, Perpignan, France
- Electrochemical Signaling in Development and Disease, Max-Delbrück-Center for Molecular Medicine (MDC), 13125, Berlin-Buch, Germany
| | - Adam Gbankoto
- Department of Animal Physiology, Faculty of Sciences and Technics, University of Abomey-Calavi, 01 BP 526, Cotonou, Benin
| | - Christophe Canal
- Institute of Modeling and Analysis in Geo-Environmental and Health (IMAGES_ESPACE-DEV), University of Perpignan Via Domitia, 66860, Perpignan, France
| | | | - Amandine Bijoux
- Institute of Modeling and Analysis in Geo-Environmental and Health (IMAGES_ESPACE-DEV), University of Perpignan Via Domitia, 66860, Perpignan, France
| | - Daniela Panáková
- Electrochemical Signaling in Development and Disease, Max-Delbrück-Center for Molecular Medicine (MDC), 13125, Berlin-Buch, Germany
| | - Anne-Cécile Ribou
- Institute of Modeling and Analysis in Geo-Environmental and Health (IMAGES_ESPACE-DEV), University of Perpignan Via Domitia, 66860, Perpignan, France.
- ESPACE-DEV, UMR UG UA UM IRD, 34093, Montpellier, France.
| |
Collapse
|
35
|
Dual subcellular compartment delivery of doxorubicin to overcome drug resistant and enhance antitumor activity. Sci Rep 2015; 5:16125. [PMID: 26530454 PMCID: PMC4632084 DOI: 10.1038/srep16125] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/09/2015] [Indexed: 01/07/2023] Open
Abstract
In order to overcome drug resistant and enhance antitumor activity of DOX, a new pH-sensitive micelle (DOX/DQA-DOX@DSPE-hyd-PEG-AA) was prepared to simultaneously deliver DOX to nucleus and mitochondria. Drug released from DOX/DQA-DOX@DSPE-hyd-PEG-AA showed a pH-dependent manner. DOX/DQA-DOX@DSPE-hyd-PEG-AA induced the depolarization of mitochondria and apoptosis in MDA-MB-231/ADR cells and A549 cells, which resulted in the high cytotoxicity of DOX/DQA-DOX@DSPE-hyd-PEG-AA against MDA-MB-231/ADR cells and A549 cells. Confocal microscopy confirmed that DOX/DQA-DOX@DSPE-hyd-PEG-AA simultaneously delivered DQA-DOX and DOX to the mitochondria and nucleus of tumor cell. After DOX/DQA-DOX@DSPE-hyd-PEG-AA was injected to the tumor-bearing nude mice by the tail vein, DOX was mainly found in tumor tissue. But DOX was widely distributed in the whole body after the administration of free DOX. Compared with free DOX, the same dose of DOX/DQA-DOX@DSPE-hyd-PEG-AA significantly inhibited the growth of DOX-resistant tumor in tumor-bearing mice without obvious systemic toxicity. Therefore, dual subcellular compartment delivery of DOX greatly enhanced the antitumor activity of DOX on DOX-resistant tumor. DOX/DQA-DOX@DSPE-hyd-PEG-AA has the potential in target therapy for DOX-resistant tumor.
Collapse
|
36
|
Zhou J, Li J, Ding X, Liu J, Luo Z, Liu Y, Ran Q, Cai K. Multifunctional Fe2O3@PPy-PEG nanocomposite for combination cancer therapy with MR imaging. NANOTECHNOLOGY 2015; 26:425101. [PMID: 26422003 DOI: 10.1088/0957-4484/26/42/425101] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
In recent years, magnetic hyperthermia nanoparticles have drawn great attention for cancer therapy because they have no limitation of tissue penetration during the therapy process. In this study, cubic nanoporous Fe2O3 nanoparticles derived from cubic Prussian blue nanoparticles were used as magnetic cores to generate heat by alternating the current magnetic field (AMF) for killing cancer cells. In addition, polypyrrole (PPy) was coated on the surfaces of the cubic Fe2O3 nanoparticles to load doxorubicin hydrochloride (DOX). The PEG component was then physically adsorbed onto the surfaces of the nanoparticles, resulting in a Fe2O3@PPy-DOX-PEG nanocomposite. The nanocomposite was triggered by acid stimulus and AMF to release DOX, resulting in a remarkable combination therapeutic effect via chemotherapy and magnetic hyperthermia. Furthermore, the nanocomposite could realize magnetic resonance imaging (MRI) due to the magnetic core structure. The study provides an alternative for the development of new nanocomposites for combination cancer therapy with MR imaging in vivo.
Collapse
Affiliation(s)
- Jun Zhou
- Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Jean SR, Tulumello DV, Riganti C, Liyanage SU, Schimmer AD, Kelley SO. Mitochondrial Targeting of Doxorubicin Eliminates Nuclear Effects Associated with Cardiotoxicity. ACS Chem Biol 2015; 10:2007-15. [PMID: 26038829 DOI: 10.1021/acschembio.5b00268] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The highly effective anticancer agent doxorubicin (Dox) is a frontline drug used to treat a number of cancers. While Dox has a high level of activity against cancer cells, its clinical use is often complicated by dose-limiting cardiotoxicity. While this side effect has been linked to the drug's direct activity in the mitochondria of cardiac cells, recent studies have shown that these result primarily from downstream effects of nuclear DNA damage. Our lab has developed a mitochondrially targeted derivative of Dox that enables the selective study of toxicity generated by the presence of Dox in the mitochondria of human cells. We demonstrate that mitochondria-targeted doxorubicin (mtDox) lacks any direct nuclear effects in H9c2 rat cardiomyocytes, and that these cells are able to undergo mitochondrial biogenesis. This recovery response compensates for the mitotoxic effects of Dox and prevents cell death in cardiomyocytes. Furthermore, cardiac toxicity was only observed in Dox but not mtDox treated mice. This study supports the hypothesis that mitochondrial damage is not the main source of the cardiotoxic effects of Dox.
Collapse
Affiliation(s)
| | | | - Chiara Riganti
- Department
of Oncology, University of Torino, via Santena 5/bis, 10126 Torino, Italy
| | | | | | - Shana O. Kelley
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| |
Collapse
|
38
|
Ismail HM, Dorchies OM, Perozzo R, Strosova MK, Scapozza L, Ruegg UT. Inhibition of iPLA2 β and of stretch-activated channels by doxorubicin alters dystrophic muscle function. Br J Pharmacol 2014; 169:1537-50. [PMID: 23849042 DOI: 10.1111/bph.12188] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 02/28/2013] [Accepted: 03/15/2013] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Chronic elevation in intracellular Ca(2+) concentration participates in death of skeletal muscle from mdx mice, a model for Duchenne muscular dystrophy (DMD). Candidate pathways mediating this Ca(2+) overload involve store-operated channels (SOCs) and stretch-activated channels (SACs), which are modulated by the Ca(2+) -independent form of PL A2 (iPLA2 ). We investigated the effect of doxorubicin (Dox), a chemotherapeutic agent reported to inhibit iPLA2 in other systems, on the activity of this enzyme and on the consequences on Ca(2+) handling and muscle function in mdx mice. EXPERIMENTAL APPROACH Effects of Dox on iPLA2 activity, reactive oxygen species production and on Ca(2+) influx were investigated in C2C12 and mdx myotubes. The mechanism of Dox-mediated iPLA2 inhibition was evaluated using purified 6x histidine-tagged enzyme. Aequorin technology was used to assess Ca(2+) concentrations underneath the plasma membrane. Isolated muscles were exposed to fatigue protocols and eccentric contractions to evaluate the effects of Dox on muscle function. KEY RESULTS Dox at 1-30 μM inhibited iPLA2 activity in cells and in the purified enzyme. Dox also inhibited SAC- but not SOC-mediated Ca(2+) influx in myotubes. Stimulated elevations of Ca(2+) concentrations below the plasmalemma were also blocked. Exposure of excised muscle to Dox was not deleterious to force production and promoted recovery from eccentric contractions. CONCLUSIONS AND IMPLICATIONS Dox showed efficacy against targets known to play a role in the pathology of DMD, namely iPLA2 and SAC. The potent SAC inhibitory effect of Dox is a novel finding that can explain partly the cardiomyopathy seen in chronic anthracycline treatment.
Collapse
Affiliation(s)
- H M Ismail
- Pharmacology, Geneva-Lausanne School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, Geneva, Switzerland
| | | | | | | | | | | |
Collapse
|
39
|
Soriani A, Iannitto ML, Ricci B, Fionda C, Malgarini G, Morrone S, Peruzzi G, Ricciardi MR, Petrucci MT, Cippitelli M, Santoni A. Reactive oxygen species- and DNA damage response-dependent NK cell activating ligand upregulation occurs at transcriptional levels and requires the transcriptional factor E2F1. THE JOURNAL OF IMMUNOLOGY 2014; 193:950-60. [PMID: 24913980 DOI: 10.4049/jimmunol.1400271] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Increasing evidence indicates that cancer cell stress induced by chemotherapeutic agents promote antitumor immune responses and contribute to their full clinical efficacy. In this article, we identify the signaling events underlying chemotherapy-induced NKG2D and DNAM-1 ligand expression on multiple myeloma (MM) cells. Our findings indicate that sublethal doses of doxorubicin and melphalan initiate a DNA damage response (DDR) controlling ligand upregulation on MM cell lines and patient-derived malignant plasma cells in Chk1/2-dependent and p53-independent manner. Drug-induced MICA and PVR gene expression are transcriptionally regulated and involve DDR-dependent E2F1 transcription factor activity. We also describe the involvement of changes in the redox state in the control of DDR-dependent upregulation of ligand surface expression and gene transcriptional activity by using the antioxidant agent N-acetyl-L-cysteine. Finally, in accordance with much evidence indicating that DDR and oxidative stress are major determinants of cellular senescence, we found that redox-dependent DDR activation upon chemotherapeutic treatment is critical for MM cell entry in premature senescence and is required for the preferential ligand upregulation on senescent cells, which are preferentially killed by NK cells and trigger potent IFN-γ production. We propose immunogenic senescence as a mechanism that promotes the clearance of drug-treated tumor cells by innate effector lymphocytes, including NK cells.
Collapse
Affiliation(s)
- Alessandra Soriani
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00161 Rome, Italy;
| | - Maria Luisa Iannitto
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00161 Rome, Italy
| | - Biancamaria Ricci
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00161 Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00161 Rome, Italy
| | - Giulia Malgarini
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00161 Rome, Italy
| | - Stefania Morrone
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Giovanna Peruzzi
- Center for Life Nano Science-Italian Institute of Technology Sapienza, 00161 Rome, Italy; and
| | - Maria Rosaria Ricciardi
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, 00161 Rome, Italy
| | - Maria Teresa Petrucci
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00161 Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, 00161 Rome, Italy;
| |
Collapse
|
40
|
Khiati S, Dalla Rosa I, Sourbier C, Ma X, Rao VA, Neckers LM, Zhang H, Pommier Y. Mitochondrial topoisomerase I (top1mt) is a novel limiting factor of doxorubicin cardiotoxicity. Clin Cancer Res 2014; 20:4873-81. [PMID: 24714774 DOI: 10.1158/1078-0432.ccr-13-3373] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Doxorubicin is one of the most effective chemotherapeutic agents. However, up to 30% of the patients treated with doxorubicin suffer from congestive heart failure. The mechanism of doxorubicin cardiotoxicity is likely multifactorial and most importantly, the genetic factors predisposing to doxorubicin cardiotoxicity are unknown. On the basis of the fact that mtDNA lesions and mitochondrial dysfunctions have been found in human hearts exposed to doxorubicin and that mitochondrial topoisomerase 1 (Top1mt) specifically controls mtDNA homeostasis, we hypothesized that Top1mt knockout (KO) mice might exhibit hypersensitivity to doxorubicin. EXPERIMENTAL DESIGN Wild-type (WT) and KO Top1mt mice were treated once a week with 4 mg/kg doxorubicin for 8 weeks. Heart tissues were analyzed one week after the last treatment. RESULTS Genetic inactivation of Top1mt in mice accentuates mtDNA copy number loss and mtDNA damage in heart tissue following doxorubicin treatment. Top1mt KO mice also fail to maintain respiratory chain protein production and mitochondrial cristae ultrastructure organization. These mitochondrial defects result in decreased O2 consumption, increased reactive oxygen species production, and enhanced heart muscle damage in animals treated with doxorubicin. Accordingly, Top1mt KO mice die within 45 days after the last doxorubicin injection, whereas the WT mice survive. CONCLUSIONS Our results provide evidence that Top1mt, which is conserved across vertebrates, is critical for cardiac tolerance to doxorubicin and adaptive response to doxorubicin cardiotoxicity. They also suggest the potential of Top1mt single-nucleotide polymorphisms testing to investigate patient susceptibility to doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Salim Khiati
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology
| | - Ilaria Dalla Rosa
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology
| | - Carole Sourbier
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute
| | - Xuefei Ma
- Laboratory of Molecular Cardiology, National Heart, Lung, and Blood Institute, NIH; and
| | - V Ashutosh Rao
- Center for Drug Evaluation and Research, Food and Drug Administration, Bethesda, Maryland
| | - Leonard M Neckers
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute
| | - Hongliang Zhang
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology;
| |
Collapse
|
41
|
Cole MP, Tangpong J, Oberley TD, Chaiswing L, Kiningham KK, St. Clair DK. Nuclear interaction between ADR-induced p65 and p53 mediates cardiac injury in iNOS (-/-) mice. PLoS One 2014; 9:e89251. [PMID: 24586632 PMCID: PMC3934890 DOI: 10.1371/journal.pone.0089251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 01/17/2014] [Indexed: 11/20/2022] Open
Abstract
Adriamycin (ADR) treatment causes an imbalance in the levels of nitric oxide (•NO) and superoxide (O2•−) production leading to cardiac injury. Previously we demonstrated that mice lacking inducible nitric oxide synthase (iNOS) have increased oxidative stress and mitochondrial injury. The molecular events leading to increased mitochondrial injury in iNOS deficient mice is unknown. ADR in the absence of iNOS preferentially activates a proapoptotic pathway without a concurrent increase in prosurvival pathways. Treatment with ADR leads to an increase in DNA binding activity of nuclear factor kappa B (NFκB) and p53 in wildtype mice. Following ADR treatment, p53, but not NFκB DNA binding activity, as well as the level of Bax, a p53 target gene, was increased in iNOS (−/−) mice. This apoptotic signaling effect in iNOS (−/−) is alleviated by overexpression of manganese superoxide dismutase (MnSOD). Increases in NFκB and p53 in ADR-treated wildtype mice did not lead to increases in target genes such as MnSOD, bcl-xL, or Bax. Moreover, co-immunoprecipitation analysis revealed that p65, a prominent member of the NFκB family, interacts with p53 in the nucleus. These results suggest that NFκB and p53 may counter act one another's actions in ADR-treated wildtype (WT) mice. Further, these results identify a novel mechanism by which oxidative stress may regulate transcription of proapoptotic genes.
Collapse
Affiliation(s)
- Marsha P. Cole
- Biochemistry and Molecular Biology, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| | - Jitbanjong Tangpong
- School of Allied Health Sciences and Public Health, Walailak University, Nakhon Si Thammarat, Thailand
| | - Terry D. Oberley
- Department of Pathology, VA Hospital, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Luksana Chaiswing
- Department of Pathology, VA Hospital, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Kinsley K. Kiningham
- Pharmaceutical, Social and Administrative Sciences, Belmont College of Pharmacy, Nashville, Tennessee, United States of America
| | - Daret K. St. Clair
- Graduate Centers for Toxicology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky, United States of America
| |
Collapse
|
42
|
Abstract
The transcription factor nuclear factor (erythroid-derived 2)-like 2, also known as NFE2L2 or NRF2, is a master regulator of the anti-oxidative stress response and positively controls the expression of a battery of anti-oxidative stress response proteins and enzymes implicated in detoxification and glutathione generation. Although its detoxifying activity is important in cancer prevention, it has recently been shown that cancer cells also exploit its protective functions to thrive and resist chemotherapy. NRF2 was also shown to the pentose phosphate pathway and glutaminolysis, which promotes purine synthesis for supporting rapid proliferation and glutathione for providing anti-oxidative stress protection. Evidence obtained from cancer patients and cell lines suggest that NRF2 is highly active in a variety of human cancers and is associated with aggressiveness. p53 is a tumor suppressor that also promotes an anti-oxidative stress metabolic program and glutaminolysis. Here we will discuss the similarities between NRF2 and p53 and review evidence that p53 might be exploited by cancer cells to gain protection against oxidative stress, as is the case for NRF2. We discuss findings of co-regulation between these transcription factors and propose possible therapeutic strategies that can be used for treatment of cancers that harbor WT p53 and express high levels of NRF2.
Collapse
|
43
|
Chamberlain GR, Tulumello DV, Kelley SO. Targeted delivery of doxorubicin to mitochondria. ACS Chem Biol 2013; 8:1389-95. [PMID: 23590228 DOI: 10.1021/cb400095v] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Several families of highly effective anticancer drugs are selectively toxic to cancer cells because they disrupt nucleic acid synthesis in the nucleus. Much less is known, however, about whether interfering with nucleic acid synthesis in the mitochondria would have significant cellular effects. In this study, we explore this with a mitochondrially targeted form of the anticancer drug doxorubicin, which inhibits DNA topoisomerase II, an enzyme that is both in mitochondria and nuclei of human cells. When doxorubicin is attached to a peptide that targets mitochondria, it exhibits significant toxicity. However, when challenged with a cell line that overexpresses a common efflux pump, it does not exhibit the reduced activity of the nuclear-localized parent drug and resists being removed from the cell. These results indicate that targeting drugs to the mitochondria provides a means to limit drug efflux and provide evidence that a mitochondrially targeted DNA topoisomerase poison is active within the organelle.
Collapse
Affiliation(s)
- Graham R. Chamberlain
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, ‡Department of Biochemistry,
Faculty of Medicine, University of Toronto, Ontario, Canada
| | - David V. Tulumello
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, ‡Department of Biochemistry,
Faculty of Medicine, University of Toronto, Ontario, Canada
| | - Shana O. Kelley
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, ‡Department of Biochemistry,
Faculty of Medicine, University of Toronto, Ontario, Canada
| |
Collapse
|
44
|
Nawara K, McCracken JL, Krysiński P, Blanchard GJ. Structure-Dependent Complexation of Fe3+ by Anthracyclines. 1. The Importance of Pendent Hydroxyl Functionality. J Phys Chem B 2013; 117:6859-67. [DOI: 10.1021/jp402349e] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Krzysztof Nawara
- Department of Chemistry, University of Warsaw, Pasteura 1, Warsaw 02-093, Poland
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - John L. McCracken
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| | - Paweł Krysiński
- Department of Chemistry, University of Warsaw, Pasteura 1, Warsaw 02-093, Poland
| | - G. J. Blanchard
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
45
|
Toita R, Murata M, Abe K, Narahara S, Piao JS, Kang JH, Ohuchida K, Hashizume M. Biological evaluation of protein nanocapsules containing doxorubicin. Int J Nanomedicine 2013; 8:1989-99. [PMID: 23717042 PMCID: PMC3662465 DOI: 10.2147/ijn.s40239] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
This study describes the applications of a naturally occurring small heat shock protein (Hsp) that forms a cage-like structure to act as a drug carrier. Mutant Hsp cages (HspG41C) were expressed in Escherichia coli by substituting glycine 41 located inside the cage with a cysteine residue to allow conjugation with a fluorophore or a drug. The HspG41C cages were taken up by various cancer cell lines, mainly through clathrin-mediated endocytosis. The cages were detected in acidic organelles (endosomes/lysosomes) for at least 48 hours, but none were detected in the mitochondria or nuclei. To generate HspG41C cages carrying doxorubicin (DOX), an anticancer agent, the HspG41C cages and DOX were conjugated using acid-labile hydrazone linkers. The release of DOX from HspG41C cages was accelerated at pH 5.0, but was negligible at pH 7.2. The cytotoxic effects of HspG41C–DOX against Suit-2 and HepG2 cells were slightly weaker than those of free DOX, but the effects were almost identical in Huh-7 cells. Considering the relatively low release of DOX from HspG41C–DOX, HspG41C–DOX exhibited comparable activity towards HepG2 and Suit-2 cells and slightly stronger cytotoxicity towards Huh-7 cells than free DOX. Hsp cages offer good biocompatibility, are easy to prepare, and are easy to modify; these properties facilitate their use as nanoplatforms in drug delivery systems and in other biomedical applications.
Collapse
Affiliation(s)
- Riki Toita
- Innovation Center for Medical Redox Navigation, Department of Advanced Medical Initiatives, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Khongkow M, Olmos Y, Gong C, Gomes AR, Monteiro LJ, Yagüe E, Cavaco TB, Khongkow P, Man EP, Laohasinnarong S, Koo CY, Harada-Shoji N, Tsang JWH, Coombes R, Schwer B, Khoo US, Lam EWF. SIRT6 modulates paclitaxel and epirubicin resistance and survival in breast cancer. Carcinogenesis 2013; 34:1476-86. [DOI: 10.1093/carcin/bgt098] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
47
|
Shaul P, Frenkel M, Goldstein EB, Mittelman L, Grunwald A, Ebenstein Y, Tsarfaty I, Fridman M. The structure of anthracycline derivatives determines their subcellular localization and cytotoxic activity. ACS Med Chem Lett 2013; 4:323-8. [PMID: 24900668 DOI: 10.1021/ml3002852] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 02/04/2013] [Indexed: 12/18/2022] Open
Abstract
The cytotoxic activities and subcellular localizations of clinically used and synthetic analogues of the anthracycline family of chemotherapeutic agents were studied. The structures of the anthracycline derivatives affected their cytotoxicity and the time required for these compounds to exert cytotoxic effects on tumor cells. Fluorescent DNA intercalator displacement experiments demonstrated that there was no correlation between the DNA intercalation properties and the cytotoxicity of the studied anthracycline derivatives. Confocal microscopy experiments indicated that structural differences led to differences in subcellular localization. All studied anthracycline derivatives were observed in lysosomes, suggesting that this organelle, which is involved in several processes leading to malignancy, may contain previously unidentified molecular targets for these antitumor agents.
Collapse
Affiliation(s)
- Pazit Shaul
- Department
of Organic Chemistry and ‡Department of Chemical Physics, School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
- Department
of Clinical Microbiology and Immunology and ∥Sackler Cellular and Molecular Imaging
Center, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Michael Frenkel
- Department
of Organic Chemistry and ‡Department of Chemical Physics, School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
- Department
of Clinical Microbiology and Immunology and ∥Sackler Cellular and Molecular Imaging
Center, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Elinor Briner Goldstein
- Department
of Organic Chemistry and ‡Department of Chemical Physics, School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
- Department
of Clinical Microbiology and Immunology and ∥Sackler Cellular and Molecular Imaging
Center, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Leonid Mittelman
- Department
of Organic Chemistry and ‡Department of Chemical Physics, School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
- Department
of Clinical Microbiology and Immunology and ∥Sackler Cellular and Molecular Imaging
Center, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Assaf Grunwald
- Department
of Organic Chemistry and ‡Department of Chemical Physics, School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
- Department
of Clinical Microbiology and Immunology and ∥Sackler Cellular and Molecular Imaging
Center, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Yuval Ebenstein
- Department
of Organic Chemistry and ‡Department of Chemical Physics, School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
- Department
of Clinical Microbiology and Immunology and ∥Sackler Cellular and Molecular Imaging
Center, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Ilan Tsarfaty
- Department
of Organic Chemistry and ‡Department of Chemical Physics, School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
- Department
of Clinical Microbiology and Immunology and ∥Sackler Cellular and Molecular Imaging
Center, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Micha Fridman
- Department
of Organic Chemistry and ‡Department of Chemical Physics, School
of Chemistry, Raymond and Beverly Sackler Faculty of Exact Sciences, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
- Department
of Clinical Microbiology and Immunology and ∥Sackler Cellular and Molecular Imaging
Center, Sackler School of Medicine, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| |
Collapse
|
48
|
Ak G, Sanlıer SH. SYNTHESIS OF FOLATE RECEPTOR-TARGETED AND DOXORUBICIN-COUPLED CHEMOTHERAPEUTIC NANOCONJUGATE AND RESEARCH INTO ITS MEDICAL APPLICATIONS. Prep Biochem Biotechnol 2012; 42:551-63. [DOI: 10.1080/10826068.2012.662926] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
49
|
Beneficial effects of curcumin on antitumor activity and adverse reactions of doxorubicin. Int J Pharm 2012; 432:42-9. [DOI: 10.1016/j.ijpharm.2012.04.062] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 04/16/2012] [Accepted: 04/23/2012] [Indexed: 11/20/2022]
|
50
|
Ak G, Yurt Lambrecht F, Sanlier SH. Radiolabeling of folate targeted multifunctional conjugate with Technetium-99m and biodistribution studies in rats. J Drug Target 2012; 20:509-14. [DOI: 10.3109/1061186x.2012.686038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|