1
|
Cetin M, Saatci O, Rezaeian AH, Rao CN, Beneker C, Sreenivas K, Taylor H, Pederson B, Chatzistamou I, Buckley B, Lessner S, Angel P, McInnes C, Sahin O. A highly potent bi-thiazole inhibitor of LOX rewires collagen architecture and enhances chemoresponse in triple-negative breast cancer. Cell Chem Biol 2024; 31:1926-1941.e11. [PMID: 39043186 PMCID: PMC11585458 DOI: 10.1016/j.chembiol.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 04/12/2024] [Accepted: 06/22/2024] [Indexed: 07/25/2024]
Abstract
Lysyl oxidase (LOX) is upregulated in highly stiff aggressive tumors, correlating with metastasis, resistance, and worse survival; however, there are currently no potent, safe, and orally bioavailable small molecule LOX inhibitors to treat these aggressive desmoplastic solid tumors in clinics. Here we discovered bi-thiazole derivatives as potent LOX inhibitors by robust screening of drug-like molecules combined with cell/recombinant protein-based assays. Structure-activity relationship analysis identified a potent lead compound (LXG6403) with ∼3.5-fold specificity for LOX compared to LOXL2 while not inhibiting LOXL1 with a competitive, time- and concentration-dependent irreversible mode of inhibition. LXG6403 shows favorable pharmacokinetic properties, globally changes ECM/collagen architecture, and reduces tumor stiffness. This leads to better drug penetration, inhibits FAK signaling, and induces ROS/DNA damage, G1 arrest, and apoptosis in chemoresistant triple-negative breast cancer (TNBC) cell lines, PDX organoids, and in vivo. Overall, our potent and tolerable bi-thiazole LOX inhibitor enhances chemoresponse in TNBC, the deadliest breast cancer subtype.
Collapse
Affiliation(s)
- Metin Cetin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ozge Saatci
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Abdol-Hossein Rezaeian
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Chintada Nageswara Rao
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Chad Beneker
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Kukkamudi Sreenivas
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Harrison Taylor
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Breanna Pederson
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Ioulia Chatzistamou
- Department of Pathology, Microbiology & Immunology, University of South Carolina, Columbia, SC 29208, USA
| | - Brian Buckley
- Small Molecule Screening Shared Resource, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Susan Lessner
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29208, USA
| | - Peggi Angel
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Bruker-MUSC Center of Excellence, Clinical Glycomics, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Campbell McInnes
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Ozgur Sahin
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
2
|
Ali T, Li D, Ponnamperumage TNF, Peterson AK, Pandey J, Fatima K, Brzezinski J, Jakusz JAR, Gao H, Koelsch GE, Murugan DS, Peng X. Generation of Hydrogen Peroxide in Cancer Cells: Advancing Therapeutic Approaches for Cancer Treatment. Cancers (Basel) 2024; 16:2171. [PMID: 38927877 PMCID: PMC11201821 DOI: 10.3390/cancers16122171] [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: 05/02/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer cells show altered antioxidant defense systems, dysregulated redox signaling, and increased generation of reactive oxygen species (ROS). Targeting cancer cells through ROS-mediated mechanisms has emerged as a significant therapeutic strategy due to its implications in cancer progression, survival, and resistance. Extensive research has focused on selective generation of H2O2 in cancer cells for selective cancer cell killing by employing various strategies such as metal-based prodrugs, photodynamic therapy, enzyme-based systems, nano-particle mediated approaches, chemical modulators, and combination therapies. Many of these H2O2-amplifying approaches have demonstrated promising anticancer effects and selectivity in preclinical investigations. They selectively induce cytotoxicity in cancer cells while sparing normal cells, sensitize resistant cells, and modulate the tumor microenvironment. However, challenges remain in achieving selectivity, addressing tumor heterogeneity, ensuring efficient delivery, and managing safety and toxicity. To address those issues, H2O2-generating agents have been combined with other treatments leading to optimized combination therapies. This review focuses on various chemical agents/approaches that kill cancer cells via H2O2-mediated mechanisms. Different categories of compounds that selectively generate H2O2 in cancer cells are summarized, their underlying mechanisms and function are elucidated, preclinical and clinical studies as well as recent advancements are discussed, and their prospects as targeted therapeutic agents and their therapeutic utility in combination with other treatments are explored. By understanding the potential of these compounds, researchers can pave the way for the development of effective and personalized cancer treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xiaohua Peng
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 3210 N. Cramer Street, Milwaukee, WI 53211, USA; (T.A.); (D.L.); (T.N.F.P.); (A.K.P.); (J.P.); (K.F.); (J.B.); (J.A.R.J.); (H.G.); (G.E.K.); (D.S.M.)
| |
Collapse
|
3
|
Defo Deeh PB, Sathiyaseelan A, Vishven Naveen K, Wang MH. Phytochemical analysis and antioxidant potential of Mondia whitei and Guibourtia tessmannii against H 2O 2-induced cytotoxicity in PC3 cells. J Biomol Struct Dyn 2024:1-15. [PMID: 38651748 DOI: 10.1080/07391102.2024.2335299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024]
Abstract
The management of oxidative stress-related disorders has garnered significant interest, particularly in the exploration of medicinal plants possessing potent antioxidant activities. This study was undertaken to evaluate the antioxidant activity of Mondia whitei (MW) and Guibourtia tessmannii (GT) against H2O2-induced cytotoxicity in PC3 cells. The phytochemical composition of MW and GT was determined by GC-MS analysis. Total phenolic (TP) and total flavonoid (TF) contents were quantified by Folin Ciocalteu and AlCl3 methods, respectively. The antioxidant potential of the extracts was determined using the DPPH and ABTS+ radicals scavenging method, as well as cupric and ferric reducing capacity assay. Moreover, all phytocompounds were docked against acetylcholinesterase (AChE) and glutathione S-transferase (GST) using ArgusLab, and results were analyzed using the BIOVIA Discovery Studio Visualizer 2021 client. MW and GT comprised 20 and 22 compounds, respectively. GT exhibited higher TP and TF contents (210.70 ± 12.7; 12.61 ± 1.3 GAE/g DW) compared to MW (132.59 ± 12.59; 5.53 ± 1.3 mg of GAE/g DW). Both MW and GT demonstrated substantial antioxidant activity, with GT proving to be more effective in preventing H2O2-induced cytotoxicity. For instance, MW and GT significantly (p < .001) increased the DPPH, ABTS+, and cupric activity, compared with the H2O2 group. All compounds identified in MW and GT exhibited a strong binding affinity against AChE and GST. Drug likeness and toxicity of all phytocompounds were under the acceptable norms of Lipinski's rule. In conclusion, these plants could be effective candidates for the management/treatment of oxidative stress-related disorders.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Patrick Brice Defo Deeh
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, Republic of Korea
| | | | - Kumar Vishven Naveen
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, Republic of Korea
| | - Myeong-Hyeon Wang
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
4
|
Selvam R, Pearl WG, Perevedentseva E, Karmenyan A, Cheng CL. Fe-doped nanodiamond-based photo-Fenton catalyst for dual-modal fluorescence imaging and improved chemotherapeutic efficacy against tumor hypoxia. RSC Adv 2024; 14:4285-4300. [PMID: 38298935 PMCID: PMC10828792 DOI: 10.1039/d3ra08465e] [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: 12/11/2023] [Accepted: 01/17/2024] [Indexed: 02/02/2024] Open
Abstract
The deficiency of oxygen in most solid tumors plays a profound role in their proliferation, metastasis, and invasion and contributes to their resistance to treatments such as radiation, chemotherapy, and photodynamic therapy (PDT). A therapeutic approach based on the Fenton reaction has received considerable interest as a means of treating cancer with ROS-based nano catalytic medicine, referred to as chemodynamic therapy (CDT). A range of modified treatment strategies are being explored to enhance both CDT and conventional methods of therapy. These include Fenton-like reactions, photo-enhanced Fenton reactions, and Fenton catalytic-enhanced synergistic therapies. In this article, we propose and demonstrate a photochemotherapy (PCT) strategy for cancer treatment utilizing near-infrared (NIR)-induced Fenton reactions using Fe-doped nanodiamond (FeND). When FeND is exposed to human lung cancer cells A549, it exhibits outstanding biocompatibility. However, when particle-treated cells are exposed to NIR laser radiation, the particle exhibits cytotoxicity to a certain degree. The anticancer medication doxorubicin (DOX) was adsorbed onto the FeND to address this issue. The conjugated DOX could undergo a redox cycle to generate excess H2O2 inside the cells, and in addition, DOX can also cause tumor cell apoptosis. Combining chemotherapy (via DOX) with a Fenton reaction results in enhanced therapeutic effectiveness. Moreover, the intrinsic fluorescence of the nanodiamond in FeND can be used to monitor the interaction of particles with cells as well as their localization, thus making it an excellent imaging probe. In our study, we found that FeND could serve as a CDT agent, biomarker, drug carrier, and potentially valuable candidate for CDT agents and contribute to the further development of more effective CDT platforms using nanodiamond.
Collapse
Affiliation(s)
- Rajakar Selvam
- Department of Physics, National Dong Hwa University Taiwan
| | | | - Elena Perevedentseva
- Department of Physics, National Dong Hwa University Taiwan
- P. N. Lebedev Institute of Physics, Russian Academy of Science Moscow Russia
| | | | | |
Collapse
|
5
|
Ray L, Ray S. Enhanced anticancer activity of siRNA and drug codelivered by anionic biopolymer: overcoming electrostatic repulsion. Nanomedicine (Lond) 2023; 18:855-874. [PMID: 37503814 DOI: 10.2217/nnm-2022-0225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Aim: To codeliver an anticancer drug (doxorubicin) and siRNA in the form of nanoparticles into CD44-overexpressing colon cancer cells (HT-29) using an anionic, amphiphilic biopolymer comprising modified hyaluronic acid (6-O-[3-hexadecyloxy-2-hydroxypropyl]-hyaluronic acid). Materials & methods: Characterization of nanoparticles was performed using dynamic light scattering, scanning electron microscopy, transmission electron microscopy, molecular docking, in vitro drug release and gel mobility assays. Detailed in vitro experiments, including a gene silencing study and western blot, were also performed. Results: A 69% knockdown of the target gene was observed, and western blot showed 5.7-fold downregulation of the target protein. The repulsive forces between siRNA and 6-O-(3-hexadecyloxy-2-hydroxypropyl)-hyaluronic acid were overcome by hydrogen bonding and hydrophobic interactions. Conclusion: The authors successfully codelivered a drug and siRNA by anionic vector.
Collapse
Affiliation(s)
- Lipika Ray
- Pharmaceutics & Pharmacokinetics Division, Council of Scientific & Industrial Research-Central Drug Research Institute, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
| | - Sutapa Ray
- Department of Chemistry, Indian Institute of Technology, Kharagpur, 721302, India
| |
Collapse
|
6
|
Recent advances and futuristic potentials of nano-tailored doxorubicin for prostate cancer therapy. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
7
|
Hydrogen Peroxide Promotes the Production of Radiation-Derived EVs Containing Mitochondrial Proteins. Antioxidants (Basel) 2022; 11:antiox11112119. [PMID: 36358489 PMCID: PMC9686922 DOI: 10.3390/antiox11112119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 12/01/2022] Open
Abstract
In spite of extensive successes, cancer recurrence after radiation treatment (RT) remains one of the significant challenges in the cure of localized prostate cancer (PCa). This study focuses on elucidating a novel adaptive response to RT that could contribute to cancer recurrence. Here, we used PC3 cell line, an adenocarcinoma from a bone metastasis and radio-resistant clone 695 cell line, which survived after total radiation dose of 66 Gy (2 Gy × 33) and subsequently regrew in nude mice after exposure to fractionated radiation at 10 Gy (2 Gy × 5). Clone 695 cells not only showed an increase in surviving fraction post-radiation but also an increase in hydrogen peroxide (H2O2) production when compared to PC3 cells. At the single cell level, confocal microscope images coupled with IMARIS rendering software demonstrate an increase in mitochondrial mass and membrane potential in clone 695 cells. Utilizing the Seahorse XF96 instrument to investigate mitochondrial respiration, clone 695 cells demonstrated a higher basal Oxygen Consumption Rate (OCR), ATP-linked OCR, and proton leak compared to PC3 cells. The elevation of mitochondrial function in clone 695 cells is accompanied by an increase in mitochondrial H2O2 production. These data suggest that H2O2 could reprogram PCa’s mitochondrial homeostasis, which allows the cancer to survive and regrow after RT. Upon exposure to RT, in addition to ROS production, we found that RT induces the release of extracellular vesicles (EVs) from PC3 cells (p < 0.05). Importantly, adding H2O2 to PC3 cells promotes EVs production in a dose-dependent manner and pre-treatment with polyethylene glycol-Catalase mitigates H2O2-mediated EV production. Both RT-derived EVs and H2O2-derived EVs carried higher levels of mitochondrial antioxidant proteins including, Peroxiredoxin 3, Glutathione Peroxidase 4 as well as mitochondrial-associated oxidative phosphorylation proteins. Significantly, adding isolated functional mitochondria 24 h prior to RT shows a significant increase in surviving fractions of PC3 cells (p < 0.05). Together, our findings reveal that H2O2 promotes the production of EVs carrying mitochondrial proteins and that functional mitochondria enhance cancer survival after RT.
Collapse
|
8
|
Mohammed DF, Madlool HA, Faris M, Shalan BH, Hasan HH, Azeez NF, Abbas FH. Harnessing inorganic nanomaterials for chemodynamic cancer therapy. Nanomedicine (Lond) 2022; 17:1891-1906. [PMID: 36647807 DOI: 10.2217/nnm-2022-0187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The most important aspect of chemodynamic therapy (CDT) is the harnessing of Fenton or Fenton-like chemistry for cancer therapy within the tumor microenvironment, which occurs because of the moderate acidity and overexpressed H2O2 in the tumor microenvironment. Hydroxyl radicals (•OH) produced within tumor cells via Fenton and Fenton-like reactions cause cancer cell death. Reactive oxygen species-mediated CDT demonstrates a desired anticancer impact without the need for external stimulation or the development of drug resistance. Cancer therapy based on CDT is known as a viable cancer therapy modality. This review discusses the most recent CDT advancements and provides some typical instances. As a result, potential methods for further improving CDT efficiency under the guidance of Fenton chemistry are offered.
Collapse
Affiliation(s)
- Dhelal F Mohammed
- Department of Pharmacy, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Hussein A Madlool
- Radiological Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Mohammed Faris
- Department of Dentistry, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Bashar Hadi Shalan
- Anesthesia Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Huda Hadi Hasan
- Department of Business Administration, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Nidaa F Azeez
- Department of Medical Physics, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Fatima Hashim Abbas
- Department of Medical Laboratory Techniques, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| |
Collapse
|
9
|
Johnson RP, Ratnacaram CK, Kumar L, Jose J. Combinatorial approaches of nanotherapeutics for inflammatory pathway targeted therapy of prostate cancer. Drug Resist Updat 2022; 64:100865. [PMID: 36099796 DOI: 10.1016/j.drup.2022.100865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PC) is the most prevalent male urogenital cancer worldwide. PC patients presenting an advanced or metastatic cancer succumb to the disease, even after therapeutic interventions including radiotherapy, surgery, androgen deprivation therapy (ADT), and chemotherapy. One of the hallmarks of PC is evading immune surveillance and chronic inflammation, which is a major challenge towards designing effective therapeutic formulations against PC. Chronic inflammation in PC is often characterized by tumor microenvironment alterations, epithelial-mesenchymal transition and extracellular matrix modifications. The inflammatory events are modulated by reactive nitrogen and oxygen species, inflammatory cytokines and chemokines. Major signaling pathways in PC includes androgen receptor, PI3K and NF-κB pathways and targeting these inter-linked pathways poses a major therapeutic challenge. Notably, many conventional treatments are clinically unsuccessful, due to lack of targetability and poor bioavailability of the therapeutics, untoward toxicity and multidrug resistance. The past decade witnessed an advancement of nanotechnology as an excellent therapeutic paradigm for PC therapy. Modern nanovectorization strategies such as stimuli-responsive and active PC targeting carriers offer controlled release patterns and superior anti-cancer effects. The current review initially describes the classification, inflammatory triggers and major inflammatory pathways of PC, various PC treatment strategies and their limitations. Subsequently, recent advancement in combinatorial nanotherapeutic approaches, which target PC inflammatory pathways, and the mechanism of action are discussed. Besides, the current clinical status and prospects of PC homing nanovectorization, and major challenges to be addressed towards the advancement PC therapy are also addressed.
Collapse
Affiliation(s)
- Renjith P Johnson
- Polymer Nanobiomaterial Research Laboratory, Nanoscience and Microfluidics Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Chandrahas Koumar Ratnacaram
- Cell Signaling and Cancer Biology Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Lalit Kumar
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Udupi, Karnataka 576 104, India
| | - Jobin Jose
- NITTE Deemed-to-be University, NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangalore 575018, India.
| |
Collapse
|
10
|
Lu B, Wang H, Shen X, Lu K, Wang H, Yuan L. Promoting gene transfection by ROS responsive silicon nanowire arrays. J Mater Chem B 2022; 10:5242-5250. [PMID: 35749073 DOI: 10.1039/d2tb00415a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The development of a fast and safe reactive oxygen species (ROS)-responsive vector is generally limited by the intracellular unstable ROS concentration, and a relatively long time is still needed for the complete intracellular release of drugs or genes induced by ROS. In this work, a gene transfection platform based on ROS-responsive silicon nanowire arrays (SN) is developed, to promote the gene transfection efficiency for several cell lines. Briefly, the surface of the ROS generating system, gold nanoparticle modified SN (SN-Au), is grafted with poly[(2-acryloyl)ethyl(p-boronic acid benzyl)diethylammonium bromide] (B-PDEAEA), an oxidation-responsive charge-reversal cationic polymer. Plasmid DNA (pDNA) bound on the surface through electrostatic interactions was directly delivered into the cells by the time the nanowires penetrate the cells. SN-Au can generate ROS under light treatment, which has an influence on the surface charge change of B-PDEAEA grafted on gold nanoparticles, realizing effective pDNA release in the cytosol for transfection. Nearly 80% of DNA released from the surface of the platform after treated with 1 mM ROS for 10 min. The transfection efficiency of the platform for several cell types was significantly enhanced after a short period of light exposure (3.2-fold for HeLa cells, 7.6-fold for L929 cells, 2.3-fold for BMSC cells and 6.2-fold for mESC cells). The platform also has good biocompatibility. Overall, our results suggest that ROS-responsive SN is a novel, efficient and safe platform for drug and gene transfection.
Collapse
Affiliation(s)
- Benben Lu
- Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, Department of polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China.
| | - Hengxiao Wang
- Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, Department of polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China.
| | - Xiang Shen
- Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, Department of polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China.
| | - Kunyan Lu
- Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, Department of polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China.
| | - Hongwei Wang
- Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, Department of polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China.
| | - Lin Yuan
- Key Lab of Health Chemistry and Molecular Diagnosis of Suzhou, Department of polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China.
| |
Collapse
|
11
|
Chaiswing L, Xu F, Zhao Y, Thorson J, Wang C, He D, Lu J, Ellingson SR, Zhong W, Meyer K, Luo W, St. Clair W, Clair DS. The RelB-BLNK Axis Determines Cellular Response to a Novel Redox-Active Agent Betamethasone during Radiation Therapy in Prostate Cancer. Int J Mol Sci 2022; 23:ijms23126409. [PMID: 35742868 PMCID: PMC9223669 DOI: 10.3390/ijms23126409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/01/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022] Open
Abstract
Aberrant levels of reactive oxygen species (ROS) are potential mechanisms that contribute to both cancer therapy efficacy and the side effects of cancer treatment. Upregulation of the non-canonical redox-sensitive NF-kB family member, RelB, confers radioresistance in prostate cancer (PCa). We screened FDA-approved compounds and identified betamethasone (BET) as a drug that increases hydrogen peroxide levels in vitro and protects non-PCa tissues/cells while also enhancing radiation killing of PCa tissues/cells, both in vitro and in vivo. Significantly, BET increases ROS levels and exerts different effects on RelB expression in normal cells and PCa cells. BET induces protein expression of RelB and RelB target genes, including the primary antioxidant enzyme, manganese superoxide dismutase (MnSOD), in normal cells, while it suppresses protein expression of RelB and MnSOD in LNCaP cells and PC3 cells. RNA sequencing analysis identifies B-cell linker protein (BLNK) as a novel RelB complementary partner that BET differentially regulates in normal cells and PCa cells. RelB and BLNK are upregulated and correlate with the aggressiveness of PCa in human samples. The RelB-BLNK axis translocates to the nuclear compartment to activate MnSOD protein expression. BET promotes the RelB-BLNK axis in normal cells but suppresses the RelB-BLNK axis in PCa cells. Targeted disruptions of RelB-BLNK expressions mitigate the radioprotective effect of BET on normal cells and the radiosensitizing effect of BET on PCa cells. Our study identified a novel RelB complementary partner and reveals a complex redox-mediated mechanism showing that the RelB-BLNK axis, at least in part, triggers differential responses to the redox-active agent BET by stimulating adaptive responses in normal cells but pushing PCa cells into oxidative stress overload.
Collapse
Affiliation(s)
- Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
- Correspondence: (L.C.); (D.S.C.)
| | - Fangfang Xu
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
| | - Yanming Zhao
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
| | - Jon Thorson
- Center for Pharmaceutical Research and Innovation, Lexington, KY 40536, USA;
- College of Pharmacy, Pharmaceutical Sciences Department, University of Kentucky, Lexington, KY 40536, USA
| | - Chi Wang
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Daheng He
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Jinpeng Lu
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Sally R. Ellingson
- Markey Biostatistics and Bioinformatics Shared Resource Facility, University of Kentucky, Lexington, KY 40536, USA; (C.W.); (D.H.); (J.L.); (S.R.E.)
| | - Weixiong Zhong
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (W.Z.); (K.M.)
| | - Kristy Meyer
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, WI 53705, USA; (W.Z.); (K.M.)
| | - Wei Luo
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA; (W.L.); (W.S.C.)
| | - William St. Clair
- Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA; (W.L.); (W.S.C.)
| | - Daret St. Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, 452 Health Sciences Research Building, Lexington, KY 40536, USA; (F.X.); (Y.Z.)
- Correspondence: (L.C.); (D.S.C.)
| |
Collapse
|
12
|
A Redoxable Mn Porphyrin, MnTnBuOE-2-PyP5+, Synergizes with Carboplatin in Treatment of Chemoresistant Ovarian Cell Line. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9664636. [PMID: 35898616 PMCID: PMC9313984 DOI: 10.1155/2022/9664636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/29/2022] [Indexed: 12/20/2022]
Abstract
We have employed a redox-active MnP (MnTnBuOE-2-PyP5+, Mn(III) meso-tetrakis (N-n-butoxyethylpyridinium-2-yl) porphyrin) frequently identified as superoxide dismutase mimic or BMX-001, to explore the redox status of normal ovarian cell in relation to two ovarian cancer cell lines: OV90 human serous ovarian cancer cell and chemotherapy-resistant OV90 cell (OVCD). We identified that OVCD cells are under oxidative stress due to high hydrogen peroxide (H2O2) levels and low glutathione peroxidase and thioredoxin 1. Furthermore, OVCD cells have increased glycolysis activity and mitochondrial respiration when compared to immortalized ovarian cells (hTER7) and parental cancer cells (OV90). Our goal was to study how ovarian cell growth depends upon the redox state of the cell; hence, we used MnP (BMX-001), a redox-active MnSOD mimetic, as a molecular tool to alter ovarian cancer redox state. Interestingly, OVCD cells preferentially uptake MnP relative to OV90 cells which led to increased inhibition of cell growth, glycolytic activity, OXPHOS, and ATP, in OVCD cells. These effects were further increased when MnP was combined with carboplatin. The effects were discussed with regard to the elevation in H2O2 levels, increased oxidative stress, and reduced Nrf2 levels and its downstream targets when cells were exposed to either MnP or MnP/carboplatin. It is significant to emphasize that MnP protects normal ovarian cell line, hTER7, against carboplatin toxicity. Our data demonstrate that the addition of MnP-based redox-active drugs may be used (via increasing excessively the oxidative stress of serous ovarian cancer cells) to improve cancer patients' chemotherapy outcomes, which develop resistance to platinum-based drugs.
Collapse
|
13
|
Doxorubicin-Loaded Metal-Organic Framework Nanoparticles as Acid-Activatable Hydroxyl Radical Nanogenerators for Enhanced Chemo/Chemodynamic Synergistic Therapy. MATERIALS 2022; 15:ma15031096. [PMID: 35161041 PMCID: PMC8838206 DOI: 10.3390/ma15031096] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/13/2022] [Accepted: 01/26/2022] [Indexed: 01/04/2023]
Abstract
Doxorubicin (DOX) is a widely used first-line antitumor agent; however, acquired drug resistance and side effects have become the main challenges to effective cancer therapy. Herein, DOX is loaded into iron-rich metal–organic framework/tannic acid (TA) nanocomplex to form a tumor-targeting and acid-activatable drug delivery system (MOF/TA-DOX, MTD). Under the acidic tumor microenvironment, MTD simultaneously releases DOX and ferrous ion (Fe2+) accompanied by degradation. Apart from the chemotherapeutic effect, DOX elevates the intracellular H2O2 levels through cascade reactions, which will be beneficial to the Fenton reaction between the Fe2+ and H2O2, to persistently produce hydroxyl radicals (•OH). Thus, MTD efficiently mediates chemodynamic therapy (CDT) and remarkably enhances the sensitivity of chemotherapy. More encouragingly, the cancer cell killing efficiency of MTD is up to ~86% even at the ultralow equivalent concentration of DOX (2.26 µg/mL), while the viability of normal cells remained >88% at the same concentration of MTD. Taken together, MTD is expected to serve as drug-delivery nanoplatforms and •OH nanogenerators for improving chemo/chemodynamic synergistic therapy and reducing the toxic side effects.
Collapse
|
14
|
An JM, Ju Y, Kim JH, Lee H, Jung Y, Kim J, Kim YJ, Kim J, Kim D. A metastasis suppressor Pt-dendrimer nanozyme for the alleviation of glioblastoma. J Mater Chem B 2021; 9:4015-4023. [PMID: 33954328 DOI: 10.1039/d1tb00425e] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nanozymes are nanostructure-based materials which mimic the enzymatic characteristics of natural enzymes. Biological applications of nanozymes have been highlighted in basic research, industry, and translational medicine as a new cutting-edge tool. In this work, and for the first time, we disclose a tumor alleviation property of a nanozyme that is made up of amine-terminated sixth-generation polyamidoamine dendrimers with encapsulated tiny platinum nanoparticles. We systematically conducted the synthesis and characterization of the dendrimer-encapsulated Pt nanoparticles (denoted Pt-dendrimer) and confirmed their enzymatic function (hydrogen peroxide (H2O2) decomposition) within various cell lines (normal, cancerous), including glioblastoma (GBM) cells. By understanding the effects of the Pt-dendrimer at the gene level, especially related to cancer cell metastasis, we have thoroughly demonstrated its ability for tumor alleviation and suppressing GBM migration, invasion, and adhesion. The present findings show great promise for the application of the nanozyme for use in GBM-related basic research as well as at clinical sites.
Collapse
Affiliation(s)
- Jong Min An
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Youngwon Ju
- Department of Chemistry, Research Institute for Basic Sciences, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Jeong Hee Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Hyein Lee
- Department of Chemistry, Research Institute for Basic Sciences, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Yuna Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Jaehoon Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Yong Jun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea. and Department of Pathology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohoon Kim
- Department of Chemistry, Research Institute for Basic Sciences, Kyung Hee University, Seoul 02447, Republic of Korea. and KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea. and KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea and Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea and Center for Converging Humanities, Kyung Hee University, Seoul 02447, Republic of Korea and Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
15
|
YÜKSEL B, DEVECİ ÖZKAN A. The Role of Citrus Nobiletin on Oxidative Stress Levels and Superoxide Dismutase Activities in Metastatic Castration-Resistant Prostate Cancer. COMMAGENE JOURNAL OF BIOLOGY 2021. [DOI: 10.31594/commagene.895415] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
16
|
Usama SM, Inagaki F, Kobayashi H, Schnermann MJ. Norcyanine-Carbamates Are Versatile Near-Infrared Fluorogenic Probes. J Am Chem Soc 2021; 143:5674-5679. [PMID: 33844539 DOI: 10.1021/jacs.1c02112] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Fluorogenic probes in the near-infrared (NIR) region have the potential to provide stimuli-dependent information in living organisms. Here, we describe a new class of fluorogenic probes based on the heptamethine cyanine scaffold, the most broadly used NIR chromophore. These compounds result from modification of heptamethine norcyanines with stimuli-responsive carbamate linkers. The resulting cyanine carbamates (CyBams) exhibit exceptional turn-ON ratios (∼170×) due to dual requirements for NIR emission: carbamate cleavage through 1,6-elimination and chromophore protonation. Illustrating their utility in complex in vivo settings, a γ-glutamate substituted CyBam was applied to imaging γ-glutamyl transpeptidase (GGT) activity in a metastatic model of ovarian cancer. Overall, CyBams have significant potential to extend the reach of fluorogenic strategies to intact tissue and live animal imaging applications.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Fuyuki Inagaki
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Hisataka Kobayashi
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Martin J Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
17
|
Meiyanto E, Zulfin U, Rahman A, Hanifa M, Utomo R, Haryanti S. Reactive oxygen species and senescence modulatory effects of rice bran extract on 4T1 and NIH-3T3 cells co-treatment with doxorubicin. Asian Pac J Trop Biomed 2021. [DOI: 10.4103/2221-1691.310204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
18
|
Vaneev AN, Gorelkin PV, Garanina AS, Lopatukhina HV, Vodopyanov SS, Alova AV, Ryabaya OO, Akasov RA, Zhang Y, Novak P, Salikhov SV, Abakumov MA, Takahashi Y, Edwards CRW, Klyachko NL, Majouga AG, Korchev YE, Erofeev AS. In Vitro and In Vivo Electrochemical Measurement of Reactive Oxygen Species After Treatment with Anticancer Drugs. Anal Chem 2020; 92:8010-8014. [PMID: 32441506 DOI: 10.1021/acs.analchem.0c01256] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In vivo monitoring of reactive oxygen species (ROS) in tumors during treatment with anticancer therapy is important for understanding the mechanism of action and in the design of new anticancer drugs. In this work, a platinized nanoelectrode is placed into a single cell for detection of the ROS signal, and drug-induced ROS production is then recorded. The main advantages of this method are the short incubation time with the drug and its high sensitivity which allows the detection of low intracellular ROS concentrations. We have shown that our new method can measure the ROS response to chemotherapy in tumor-bearing mice in real-time. ROS levels were measured in vivo inside the tumor at different depths in response to doxorubicin. This work provides an effective new approach for the measurement of intracellular ROS by platinized nanoelectrodes.
Collapse
Affiliation(s)
- Alexander N Vaneev
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia.,Lomonosov Moscow State University, Chemistry Department, Leninskie Gory, 1, 3, Moscow 119991, Russia
| | - Petr V Gorelkin
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia
| | - Anastasiia S Garanina
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia
| | - Helena V Lopatukhina
- Lomonosov Moscow State University, Chemistry Department, Leninskie Gory, 1, 3, Moscow 119991, Russia
| | - Stepan S Vodopyanov
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia.,Lomonosov Moscow State University, Chemistry Department, Leninskie Gory, 1, 3, Moscow 119991, Russia
| | - Anna V Alova
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia.,Lomonosov Moscow State University, Chemistry Department, Leninskie Gory, 1, 3, Moscow 119991, Russia
| | - Oxana O Ryabaya
- N. N. Blokhin National Medical Research Center of Oncology, 24 Kashirskoe shosse, Moscow 115478, Russia
| | - Roman A Akasov
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia.,I. M. Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, Moscow 119991, Russia
| | - Yanjun Zhang
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China.,Imperial College London, Department of Medicine, London W12 0NN, United Kingdom
| | - Pavel Novak
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia.,Imperial College London, Department of Medicine, London W12 0NN, United Kingdom
| | - Sergey V Salikhov
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia
| | - Maxim A Abakumov
- N. I. Pirogov Russian National Research Medical University, Ostrovityanova Street 1/7, Moscow 117997, Russia
| | - Yasufumi Takahashi
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | | | - Natalia L Klyachko
- Lomonosov Moscow State University, Chemistry Department, Leninskie Gory, 1, 3, Moscow 119991, Russia
| | - Alexander G Majouga
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia.,Lomonosov Moscow State University, Chemistry Department, Leninskie Gory, 1, 3, Moscow 119991, Russia.,D. Mendeleev University of Chemical Technology of Russia, Miusskaya Square, 9, Moscow 125047, Russia
| | - Yuri E Korchev
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia.,Imperial College London, Department of Medicine, London W12 0NN, United Kingdom
| | - Alexander S Erofeev
- National University of Science and Technology "MISiS", Leninskiy Avenue, 4, Moscow 119049, Russia.,Lomonosov Moscow State University, Chemistry Department, Leninskie Gory, 1, 3, Moscow 119991, Russia
| |
Collapse
|
19
|
An H, Guo C, Li D, Liu R, Xu X, Guo J, Ding J, Li J, Chen W, Zhang J. Hydrogen Peroxide-Activatable Nanoparticles for Luminescence Imaging and In Situ Triggerable Photodynamic Therapy of Cancer. ACS APPLIED MATERIALS & INTERFACES 2020; 12:17230-17243. [PMID: 32193923 DOI: 10.1021/acsami.0c01413] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Abnormally increased reactive oxygen species (ROS) are intimately related to the development and metastasis of cancer. Since hydrogen peroxide (H2O2) is a major component of ROS, molecular imaging and selective treatment in response to high H2O2 are intriguing for the management of cancers. Herein, we report novel self-assembly luminescent nanoparticles, which can be activated by H2O2, thereby serving as an effective nanotheranostics for luminescence imaging and in situ photodynamic therapy (PDT) of tumors with high H2O2. This functional nanomedicine was assembled from an amphiphilic conjugate (defined as CLP) based on chlorin e6 (Ce6) simultaneously conjugated with luminol and poly(ethylene glycol), exhibiting a well-defined core-shell nanostructure. Upon triggering by pathologically relevant levels of H2O2, CLP nanoparticles produced luminescence due to the luminol unit and simultaneous excitation of Ce6 by chemiluminescence resonance energy transfer, enabling in vitro and in vivo imaging of tumors with highly expressed H2O2. In addition, excited Ce6 can produce singlet oxygen (1O2) for in situ PDT of H2O2-high tumors and inhibiting lung metastasis, which was demonstrated by in vitro and in vivo experiments. Furthermore, preliminary studies revealed the biosafety of CLP nanoparticles. Consequently, the self-illuminating nanoparticles are promising for noninvasive imaging and therapy of tumors with high expression of H2O2.
Collapse
Affiliation(s)
- Huijie An
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
- Department of Pharmacy, General Hospital of Southern Theatre Command, PLA, Guangzhou 510010, China
| | - Chunhua Guo
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Dandan Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Renfeng Liu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xiaoqiu Xu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiawei Guo
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jun Ding
- Department of Ultrasound, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianjun Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wei Chen
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
20
|
Li M, Lv J, Wang S, Wang J, Lin Y. Expanded mesoporous silica-encapsulated ultrasmall Pt nanoclusters as artificial enzymes for tracking hydrogen peroxide secretion from live cells. Anal Chim Acta 2020; 1104:180-187. [PMID: 32106950 DOI: 10.1016/j.aca.2020.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/07/2020] [Indexed: 01/09/2023]
Abstract
Design of synthetic structures that possess the similar functions to natural enzymes held great promise in environmental detection and biomedical application. Herein, a new concept for the fabrication of solid-supported catalysts as peroxidase mimic have been proposed to realize high-catalytic activity and stability by utilizing expanded mesoporous silica (EMSN)-encapsulated Pt nanoclusters. Compared with PtNCs, the introduction of amino group modified EMSN would enrich H2O2 on the surface of PtNCs and increase the catalytic sites for H2O2 decomposition, which gave rise to the higher catalytic activity of EMSN-PtNCs over a broad pH range, especially in weakly acidic and neural solutions. This would facilitate their applications for real-time monitoring the secretion of H2O2 from living cancer cells stimulated by various anticancer drugs. Our findings not only pave the way to use porous matrix as the structural component for the design of the biomimetic catalysts, but also provide a simple and reliable platform to monitor H2O2 released from living cells in real time, which holds great potential for elucidating the biological roles of H2O2 and underlying molecular mechanisms of drug cytotoxicity as well as drug therapeutic effects.
Collapse
Affiliation(s)
- Meng Li
- College of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jie Lv
- College of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, 050017, China
| | - Shuangling Wang
- College of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jing Wang
- College of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, 050017, China.
| | - Yulong Lin
- College of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
21
|
Falls-Hubert KC, Butler AL, Gui K, Anderson M, Li M, Stolwijk JM, Rodman SN, Solst SR, Tomanek-Chalkley A, Searby CC, Sheffield VC, Sandfort V, Schmidt H, McCormick ML, Wels BR, Allen BG, Buettner GR, Schultz MK, Spitz DR. Disulfiram causes selective hypoxic cancer cell toxicity and radio-chemo-sensitization via redox cycling of copper. Free Radic Biol Med 2020; 150:1-11. [PMID: 32032663 PMCID: PMC7299833 DOI: 10.1016/j.freeradbiomed.2020.01.186] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/30/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023]
Abstract
Therapies for lung cancer patients initially elicit desirable responses, but the presence of hypoxia and drug resistant cells within tumors ultimately lead to treatment failure. Disulfiram (DSF) is an FDA approved, copper chelating agent that can target oxidative metabolic frailties in cancer vs. normal cells and be repurposed as an adjuvant to cancer therapy. Clonogenic survival assays showed that DSF (50-150 nM) combined with physiological levels of Cu (15 μM CuSO4) was selectively toxic to H292 NSCLC cells vs. normal human bronchial epithelial cells (HBEC). Furthermore, cancer cell toxicity was exacerbated at 1% O2, relative to 4 or 21% O2. This selective toxicity of DSF/Cu was associated with differential Cu ionophore capabilities. DSF/Cu treatment caused a >20-fold increase in cellular Cu in NSCLCs, with nearly two-fold higher Cu present in NSCLCs vs. HBECs and in cancer cells at 1% O2vs. 21% O2. DSF toxicity was shown to be dependent on the retention of Cu as well as oxidative stress mechanisms, including the production of superoxide, peroxide, lipid peroxidation, and mitochondrial damage. DSF was also shown to selectively (relative to HBECs) enhance radiation and chemotherapy-induced NSCLC killing and reduce radiation and chemotherapy resistance in hypoxia. Finally, DSF decreased xenograft tumor growth in vivo when combined with radiation and carboplatin. These results support the hypothesis that DSF could be a promising adjuvant to enhance cancer therapy based on its apparent ability to selectively target fundamental differences in cancer cell oxidative metabolism.
Collapse
Affiliation(s)
- Kelly C Falls-Hubert
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Aimee L Butler
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Kai Gui
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Michael Anderson
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Mengshi Li
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jeffrey M Stolwijk
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Samuel N Rodman
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Shane R Solst
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Ann Tomanek-Chalkley
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Charles C Searby
- Department Pediatrics, University of Iowa, Iowa City, IA, 52242, USA; Department Ophthalmology, University of Iowa, Iowa City, IA, 52242, USA
| | - Val C Sheffield
- Department Pediatrics, University of Iowa, Iowa City, IA, 52242, USA; Department Ophthalmology, University of Iowa, Iowa City, IA, 52242, USA
| | - Vanessa Sandfort
- Gastroenterology and Hepatology, Münster University Hospital (UKM), Münster, Germany
| | - Hartmut Schmidt
- Gastroenterology and Hepatology, Münster University Hospital (UKM), Münster, Germany
| | - Michael L McCormick
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Brian R Wels
- State Hygienic Lab, University of Iowa, Ankeny, IA, 50023, USA
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Garry R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Michael K Schultz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA; Department of Radiology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA; Department Pediatrics, University of Iowa, Iowa City, IA, 52242, USA; Department of Chemistry, University of Iowa, Iowa City, IA, 52241, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
22
|
Ray L. Synergistic anticancer activity by co-delivered nanosized dual therapeutic agents and siRNA in colon cancer. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101351] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
23
|
A novel metadherinΔ7 splice variant enhances triple negative breast cancer aggressiveness by modulating mitochondrial function via NFĸB-SIRT3 axis. Oncogene 2019; 39:2088-2102. [PMID: 31806873 DOI: 10.1038/s41388-019-1126-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/15/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022]
Abstract
Metadherin (MTDH) expression inversely correlates with prognosis of several cancers including mammary carcinomas. In this work, we identified a novel splice variant of MTDH with exon7 skipping (MTDHΔ7) and its levels were found significantly high in triple negative breast cancer (TNBC) cells and in patients diagnosed with TNBC. Selective overexpression of MTDHΔ7 in MDA-MB-231 and BT-549 cells enhanced proliferation, invasion, and epithelial-to-mesenchymal (EMT) transition markers in comparison to its wildtype counterpart. In contrast, knockdown of MTDHΔ7 induced antiproliferative/antiinvasive effects. Mechanistically, MTDH-NFĸB-p65 complex activated SIRT3 transcription by binding to its promoter that in turn enhanced MnSOD levels and promoted EMT in TNBC cells. Intriguingly, mitochondrial OCR through Complex-I and -IV, and glycolytic rate (ECAR) were significantly high in MDA-MB-231 cells stably expressing MTDHΔ7. While depletion of SIRT3 inhibited MTDH-Wt/Δ7-induced OCR and ECAR, knockdown of MnSOD inhibited only ECAR. In addition, MTDH-Wt/Δ7-mediated pro-proliferative/-invasive effects were greatly obviated with either siSIRT3 or siMnSOD in these cells. The functional relevance of MTDHΔ7 was further proved under in vivo conditions in an orthotopic mouse model of breast cancer. Mice bearing labeled MDA-MB-231 cells stably expressing MTDHΔ7 showed significantly more tumor growth and metastatic ability to various organs in comparison to MTDH-Wt bearing mice. Taken together, MTDHΔ7 promotes TNBC aggressiveness through enhanced mitochondrial biogenesis/function, which perhaps serves as a biomarker.
Collapse
|
24
|
Faustova M, Nikolskaya E, Sokol M, Zabolotsky A, Mollaev M, Zhunina O, Fomicheva M, Lobanov A, Severin E, Yabbarov N. High-effective reactive oxygen species inducer based on Mn-tetraphenylporphyrin loaded PLGA nanoparticles in binary catalyst therapy. Free Radic Biol Med 2019; 143:522-533. [PMID: 31520768 DOI: 10.1016/j.freeradbiomed.2019.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 01/09/2023]
Abstract
The mechanisms of binary catalyst therapy (BCT) and photodynamic therapy (PDT) are based on the formation of reactive oxygen species (ROS). This ROS formation results from specific chemical reactions. In BCT, light exposure does not necessarily initiate ROS formation and BCT application is not limited to regions of tissues that are accessible to illumination like photodynamic therapy (PDT). The principle of BCT is electron transition, resulting in the interaction of a transition metal complex (catalyst) and substrate molecule. MnIII- tetraphenylporphyrin chloride (MnClTPP) in combination with an ascorbic acid (AA) has been proposed as an appropriate candidate for cancer treatment regarding the active agents in BCT. The goal of this study was to determine whether MnClTPP in combination with AA would be a promising agent for BCT. The problem of used MnClTPP's, low solubility in water, was solved by MnClTPP loading into PLGA matrix. H2O2 produced during AA decomposition oxidized MnClTPP to high-reactive oxo-MnV species. MnClTPP in presence AA leads to the production of excessive ROS levels in vitro. ROS are mainly substrates of catalase and superoxide dismutase (H2O2 and O2●-). SOD1 and catalase were identified as the key players of the MnClTPP ROS-induced cell defense system. The cytotoxicity of MnClTPP-loaded nanoparticles (NPs) was greatly increased in the presence of specific catalase inhibitor (3-amino-1,2,4-triazole (3AT)) and superoxide dismutase 1 (SOD1) inhibitor (diethyldithiocarbamate (DDC)). Cell death resulted from the combined activation of caspase-dependent (caspase 3/9 system) and independent pathways, namely the AIF translocation to nuclei. Preliminary acute toxicity and in vivo anticancer studies have been revealed the safe and potent anticancer effect of PLGA-entrapped MnClTPP in combination with AA. The findings indicate that MnClTPP-loaded PLGA NPs are promising agents for BCT.
Collapse
Affiliation(s)
- Maria Faustova
- MIREA, Russian Technological University, Lomonosov Institute of Fine Chemical Technologies, 119454, Moscow, Russia
| | | | - Maria Sokol
- JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149, Moscow, Russia
| | | | - Murad Mollaev
- MIREA, Russian Technological University, Lomonosov Institute of Fine Chemical Technologies, 119454, Moscow, Russia
| | - Olga Zhunina
- Semenov Institute of Chemical Physics, 119991, Moscow, Russia
| | - Margarita Fomicheva
- JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149, Moscow, Russia
| | - Anton Lobanov
- Semenov Institute of Chemical Physics, 119991, Moscow, Russia
| | - Evgeniy Severin
- JSC Russian Research Center for Molecular Diagnostics and Therapy, 117149, Moscow, Russia
| | - Nikita Yabbarov
- Semenov Institute of Chemical Physics, 119991, Moscow, Russia.
| |
Collapse
|
25
|
Porter MR, Walker JM, Zaleski JM. The Outliers: Metal-Mediated Radical Reagents for Biological Substrate Degradation. Acc Chem Res 2019; 52:1957-1967. [PMID: 31243967 PMCID: PMC9573769 DOI: 10.1021/acs.accounts.9b00185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The predictable and controllable interaction of small organic or peptidic molecules with biological substrates is the primary reason most pharmaceuticals are narrowly decorated carbon frameworks. The inhibition or activation binding models are measurable and without side reactions that can cause pathological angst. Yet many diseases, especially those involving rapid proliferation of cells (i.e., cancer) or aggregation of peptides (e.g., heart disease, Alzheimer's disease) have not yet been cured by inhibition therapeutics. Additionally, interventional medicine is often required to alleviate such maladies by physical removal first, followed by molecular-level therapy as a second stage. Thus, there appears to be a niche for more aggressive therapeutics that may employ harsher chemical processes to realize clinical efficacy, albeit without causing catastrophic side effects. Molecules that may be considered for this challenge are not typically biomimetic, nor do they fit the traditional pharmaceutical paradigm. They may have unusual modes of action or undesired reactivity that can be lethal if not controlled. These are the outliers; potential pharmacophores that biology does not know how to manage or adapt to. This is why they may be an intriguing class of agents that needs continuous development. In this Account, we connect the under-developed enediyne family of compounds and our metalloenediyne derivatives to existing radical-based therapeutics such as bleomycin and doxorubicin to illustrate that controlled diradical reactivity, although an outlier mechanism, has a place in the therapeutic portfolio. This is self-evident in that of the 11 natural product enediynes known, 2 have clinical impact, a strong ratio. We expand on the chemical diversity of potential enediyne constructs and focus on the accessible trigger mechanisms to activate diradical formation as a method to control toxicity. Moreover, we further illustrate how electromagnetic fields can be employed to activate both molecular and larger nanomaterial constructs that carry highly concentrated payloads of reactive reagent. Finally, we describe how controlled diradical reactivity can reach beyond traditional therapeutic targets such as DNA, to peptide aggregates found in blood clots, neural fibrils, and membrane scaffolds. It is our belief that cleverly constructed frameworks with well-designed and controlled activation/reaction schemes can lead to novel therapeutics that can challenge evolving viral and bacterial invaders. From this evangelical perspective, our hope is that the conceptual framework, if not the specific designs in this Account, stimulate the readership to develop out-of-the-box therapeutic designs that may combat resistant disease targets.
Collapse
Affiliation(s)
- Meghan R. Porter
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Joan M. Walker
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jeffrey M. Zaleski
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
26
|
Bhattacharya S, Gong X, Wang E, Dutta SK, Caplette JR, Son M, Nguyen FT, Strano MS, Mukhopadhyay D. DNA-SWCNT Biosensors Allow Real-Time Monitoring of Therapeutic Responses in Pancreatic Ductal Adenocarcinoma. Cancer Res 2019; 79:4515-4523. [PMID: 31292162 DOI: 10.1158/0008-5472.can-18-3337] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 03/15/2019] [Accepted: 07/03/2019] [Indexed: 01/05/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly desmoplastic cancer with limited treatment options. There is an urgent need for tools that monitor therapeutic responses in real time. Drugs such as gemcitabine and irinotecan elicit their therapeutic effect in cancer cells by producing hydrogen peroxide (H2O2). In this study, specific DNA-wrapped single-walled carbon nanotubes (SWCNT), which precisely monitor H2O2, were used to determine the therapeutic response of PDAC cells in vitro and tumors in vivo. Drug therapeutic efficacy was evaluated in vitro by monitoring H2O2 differences in situ using reversible alteration of Raman G-bands from the nanotubes. Implantation of the DNA-SWCNT probe inside the PDAC tumor resulted in approximately 50% reduction of Raman G-band intensity when treated with gemcitabine versus the pretreated tumor; the Raman G-band intensity reversed to its pretreatment level upon treatment withdrawal. In summary, using highly specific and sensitive DNA-SWCNT nanosensors, which can determine dynamic alteration of hydrogen peroxide in tumor, can evaluate the effectiveness of chemotherapeutics. SIGNIFICANCE: A novel biosensor is used to detect intratumoral hydrogen peroxide, allowing real-time monitoring of responses to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, Florida.,Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Jacksonville, Florida
| | - Xun Gong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Enfeng Wang
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, Florida
| | - Shamit K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, Florida
| | - Joseph R Caplette
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Jacksonville, Florida
| | - Manki Son
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Freddy T Nguyen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Michael S Strano
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, Florida. .,Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Jacksonville, Florida
| |
Collapse
|
27
|
Izadi A, Meshkini A, Entezari MH. Mesoporous superparamagnetic hydroxyapatite nanocomposite: A multifunctional platform for synergistic targeted chemo-magnetotherapy. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 101:27-41. [PMID: 31029320 DOI: 10.1016/j.msec.2019.03.066] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/18/2022]
Abstract
In the present study, the aim was to develop a magneto-responsive nanocomposite for application in drug delivery by the integration of magnetic nanoparticles into an inorganic architecture, hydroxyapatite. The magnetic mesoporous hydroxyapatite nanocomposites, MMHAPs, were synthesized using a template-free method and fully characterized by XRD, FT-IR, TEM, FE-SEM, VSM, ICP, BET, and UV-Vis spectroscopy. MMHAPs exhibited a rod-like shape with a structure of large mesopores and high surface area. A sample of the nanocomposites with well-defined properties, MMHAP(2), was selected as a carrier for delivery of chemotherapy drug, doxorubicin (Dox). Then, it was coated with polyethylene glycol (P) and folic acid (F), providing aqueous stability and tumor targeting, respectively. The evaluation of drug release profile revealed that the release of drug occurs in a time-staggered manner under low pH conditions, which simulate the internal condition of lysosome. More important, a significant drug release was observed under a static magnetic field (SMF), displaying a magnetically triggered release. According to the toxicity assessment, MMHAP(2) did not show any noticeable toxic effect against the tumor cells (Saos-2) and normal cells (HEK-293) up to 100 μg ml-1 in the presence or absence of SMF. In contrast, the drug-loaded nanocomposite, F.P.D@MMHAP(2), possesses high antitumor efficacy particularly in the presence of SMF. Moreover, it was found that the cellular internalization of F.P.D@MMHAP(2) could be increased by SMF, providing therapeutic efficiency enhancement. The high cytotoxic effect of F.P.D@MMHAP(2) with the help of SMF caused apoptosis in the tumor cells, which was preceded by a disturbance in the intracellular redox state and then caspase activation. Based on the data obtained, F.P.D@MMHAP(2) is a pH- and magneto-responsive platform opening up a new perspective in terms of its exploitation in cancer therapy.
Collapse
Affiliation(s)
- Azadeh Izadi
- Biochemical Research Center, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Azadeh Meshkini
- Biochemical Research Center, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Mohammad H Entezari
- Sonochemical Research Center, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Environmental Chemistry Research Center, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
28
|
Renu K, Valsala Gopalakrishnan A. Deciphering the molecular mechanism during doxorubicin-mediated oxidative stress, apoptosis through Nrf2 and PGC-1α in a rat testicular milieu. Reprod Biol 2019; 19:22-37. [PMID: 30827825 DOI: 10.1016/j.repbio.2019.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 02/04/2019] [Accepted: 02/07/2019] [Indexed: 12/13/2022]
Abstract
Doxorubicin is an extensively applied anti-cancerous drug since 1950's and its usage is constrained because of its accumulation in a non-cancerous organ. Many studies have proven that doxorubicin causes reproductive toxicity depends on its dosage, particularly due to increased oxidative stress and apoptosis. A number of the researches have been carried out concerning its prevention. But there is a need to recognize the mechanism at the back of its toxicity to get better and improved method of treatment. To clarify the feasible mechanism of doxorubicin-mediated reproductive toxicity in rats, we have administrated doxorubicin at distinct dosages inclusive of low dosage (male rats that are at 230-250 g acquired cumulatively 1.5 mg/kg; ip; once per week for five weeks) and high dosage (male rats which are at 230-250 grams obtained cumulatively 15 mg/kg; ip; once every week for five weeks). Doxorubicin decreases antioxidant level such as GSH, Cu/Zn SOD, Mn SOD both in serum and testes. Increased oxidative stress is considered via elevated MDA level both in serum and testes. The level of ROS is measured via the DCFDA method in testes. Apoptosis become found through DNA fragmentation assay and quantification of Caspase 3, Caspase 9, Bcl2 and Cytochrome C. Doxorubicin mediated oxidative stress and apoptosis in testicular milieu is through deregulation of Nrf2, PGC-1α, AHR, ARNT, PXR, SUMO-1, UCP2, UCP3, ANX A5, Caspase 3, Caspase 9, Bcl2, Cytochrome C, GR, and GPX. In end, doxorubicin-mediated oxidative stress and apoptosis is through diverse transcriptional factors and genes with respect to decreased antioxidant level, augmented ROS level and Annexin A5 in the testicular milieu.
Collapse
Affiliation(s)
- Kaviyarasi Renu
- Department of Biomedical Sciences, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu, 632014, India
| | | |
Collapse
|
29
|
Chaiswing L, St. Clair WH, St. Clair DK. Redox Paradox: A Novel Approach to Therapeutics-Resistant Cancer. Antioxid Redox Signal 2018; 29:1237-1272. [PMID: 29325444 PMCID: PMC6157438 DOI: 10.1089/ars.2017.7485] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 01/05/2018] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Cancer cells that are resistant to radiation and chemotherapy are a major problem limiting the success of cancer therapy. Aggressive cancer cells depend on elevated intracellular levels of reactive oxygen species (ROS) to proliferate, self-renew, and metastasize. As a result, these aggressive cancers maintain high basal levels of ROS compared with normal cells. The prominence of the redox state in cancer cells led us to consider whether increasing the redox state to the condition of oxidative stress could be used as a successful adjuvant therapy for aggressive cancers. Recent Advances: Past attempts using antioxidant compounds to inhibit ROS levels in cancers as redox-based therapy have met with very limited success. However, recent clinical trials using pro-oxidant compounds reveal noteworthy results, which could have a significant impact on the development of strategies for redox-based therapies. CRITICAL ISSUES The major objective of this review is to discuss the role of the redox state in aggressive cancers and how to utilize the shift in redox state to improve cancer therapy. We also discuss the paradox of redox state parameters; that is, hydrogen peroxide (H2O2) as the driver molecule for cancer progression as well as a target for cancer treatment. FUTURE DIRECTIONS Based on the biological significance of the redox state, we postulate that this system could potentially be used to create a new avenue for targeted therapy, including the potential to incorporate personalized redox therapy for cancer treatment.
Collapse
Affiliation(s)
- Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky-Lexington, Lexington, Kentucky
| | - William H. St. Clair
- Department of Radiation Medicine, University of Kentucky-Lexington, Lexington, Kentucky
| | - Daret K. St. Clair
- Department of Toxicology and Cancer Biology, University of Kentucky-Lexington, Lexington, Kentucky
| |
Collapse
|
30
|
Sharma AK, Pandey S, Sharma KH, Nerthigan Y, Khan MS, Hang DR, Wu HF. Two dimensional α-MoO 3-x nanoflakes as bare eye probe for hydrogen peroxide in biological fluids. Anal Chim Acta 2018. [DOI: 10.1016/j.aca.2018.01.057] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
31
|
AbouAitah K, Swiderska-Sroda A, Farghali AA, Wojnarowicz J, Stefanek A, Gierlotka S, Opalinska A, Allayeh AK, Ciach T, Lojkowski W. Folic acid-conjugated mesoporous silica particles as nanocarriers of natural prodrugs for cancer targeting and antioxidant action. Oncotarget 2018; 9:26466-26490. [PMID: 29899871 PMCID: PMC5995188 DOI: 10.18632/oncotarget.25470] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/01/2018] [Indexed: 12/22/2022] Open
Abstract
Naturally derived prodrugs have a wide range of pharmacological activities, including anticancer, antioxidant, and antiviral effects. However, significant barriers inhibit their use in medicine, e.g. their hydrophobicity. In this comprehensive study, we investigated simple and effective nanoformulations consisting of amine-functionalized and conjugated with folic acid (FA) mesoporous silica nanoparticles (MSNs). Two types of MSNs were studied: KCC- 1, with mean size 324 nm and mean pore diameter 3.4 nm, and MCM - 41, with mean size 197 and pore diameter 2 nm. Both types of MSNs were loaded with three anticancer prodrugs: curcumin, quercetin, and colchicine. The nanoformulations were tested to target in vitro human hepatocellular carcinoma cells (HepG2) and HeLa cancer cells. The amine-functionalized and FA-conjugated curcumin-loaded, especially KCC-1 MSNs penetrated all cells organs and steadily released curcumin. The FA-conjugated MSNs displayed higher cellular uptake, sustained intracellular release, and cytotoxicity effects in comparison to non-conjugated MSNs. The KCC-1 type MSNs carrying curcumin displayed the highest anticancer activity. Apoptosis was induced through specific signaling molecular pathways (caspase-3, H2O2, c-MET, and MCL-1). The nanoformulations displayed also an enhanced antioxidant activity compared to the pure forms of the prodrugs, and the effect depended on the time of release, type of MSN, prodrug, and assay used. FA-conjugated MSNs carrying curcumin and other safe natural prodrugs offer new possibilities for targeted cancer therapy.
Collapse
Affiliation(s)
- Khaled AbouAitah
- Department of Medicinal and Aromatic Plants Research, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), Dokki, Giza, Egypt
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Swiderska-Sroda
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Ahmed A. Farghali
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef, Egypt
| | - Jacek Wojnarowicz
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Agata Stefanek
- Biomedical Engineering Laboratory, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Stanislaw Gierlotka
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Opalinska
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Abdou K. Allayeh
- Environmental Virology Laboratory, National Research Centre (NRC), Dokki, Giza, Egypt
| | - Tomasz Ciach
- Biomedical Engineering Laboratory, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Witold Lojkowski
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
32
|
Eghbal MA, Yusefi E, Tavakoli-Ardakani M, Ramazani M, Zarei MH, Salimi A, Pourahmad J. Exposure to Antineoplastic Agents Induces Cytotoxicity in Nurse Lymphocytes: Role of Mitochondrial Damage and Oxidative Stress. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2018; 17:43-52. [PMID: 29796028 PMCID: PMC5958323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cytotoxicity and mitochondrial parameters were studied in isolated lymphocytes and their mitochondria obtained from occupationally exposed nurses through inhalation exposure to antineoplastic drugs and results were compared to those of unexposed nurses. The group of occupationally exposed nurses consisted of 50 individuals ranging in age from 30 to 35 years. The control group included 50 nurses who were not occupationally exposed to the preparation and handling of antineoplastic drugs and their anthropometric and biochemical characteristics were similar to those of the expose group. All cytotoxicity and mitochondrial parameters evaluated in exposed group were significantly increased (P < 0.05) compared to the unexposed control group. Finally, the results of our study suggest that using antioxidant, mitochondrial and lysosomal protective agents can be promising drug candidates for the hospital staff in the risk of exposure to exposure to antineoplastic drugs.
Collapse
Affiliation(s)
- Mohmmad Ali Eghbal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tehran, Iran.
| | - Elham Yusefi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tehran, Iran.
| | - Maria Tavakoli-Ardakani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maral Ramazani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Hadi Zarei
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Science, Ardabil, Iran.,Corresponding authors: E-mail: ;
| | - Jalal Pourahmad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran. ,Corresponding authors: E-mail: ;
| |
Collapse
|
33
|
Oxidative stress regulates cellular bioenergetics in esophageal squamous cell carcinoma cell. Biosci Rep 2017; 37:BSR20171006. [PMID: 29026004 PMCID: PMC5725616 DOI: 10.1042/bsr20171006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/01/2017] [Accepted: 10/02/2017] [Indexed: 02/07/2023] Open
Abstract
The aim of the present study was to explore the effects of oxidative stress induced by CoCl2 and H2O2 on the regulation of bioenergetics of esophageal squamous cell carcinoma (ESCC) cell line TE-1 and analyze its underlying mechanism. Western blot results showed that CoCl2 and H2O2 treatment of TE-1 cells led to significant reduction in mitochondrial respiratory chain complex subunits expression and increasing intracellular reactive oxygen species (ROS) production. We further found that TE-1 cells treated with CoCl2, a hypoxia-mimicking reagent, dramatically reduced the oxygen consumption rate (OCR) and increased the extracellular acidification rate (ECAR). However, H2O2 treatment decreased both the mitochondrial respiration and aerobic glycolysis significantly. Moreover, we found that H2O2 induces apoptosis in TE-1 cells through the activation of PARP, Caspase 3, and Caspase 9. Therefore, our findings indicate that CoCl2 and H2O2 could cause mitochondrial dysfunction by up-regulation of ROS and regulating the cellular bioenergy metabolism, thus affecting the survival of tumor cells.
Collapse
|
34
|
Kankala RK, Liu CG, Chen AZ, Wang SB, Xu PY, Mende LK, Liu CL, Lee CH, Hu YF. Overcoming Multidrug Resistance through the Synergistic Effects of Hierarchical pH-Sensitive, ROS-Generating Nanoreactors. ACS Biomater Sci Eng 2017; 3:2431-2442. [DOI: 10.1021/acsbiomaterials.7b00569] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Ranjith Kumar Kankala
- Institute
of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Xiamen 361021, P. R. China
| | - Chen-Guang Liu
- Institute
of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Ai-Zheng Chen
- Institute
of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Xiamen 361021, P. R. China
| | - Shi-Bin Wang
- Institute
of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
- Fujian Provincial Key Laboratory of Biochemical Technology, Xiamen 361021, P. R. China
| | - Pei-Yao Xu
- Institute
of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, P. R. China
| | - Lokesh Kumar Mende
- Department
of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan
| | - Chen-Lun Liu
- Department
of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan
| | - Chia-Hung Lee
- Department
of Life Science and Institute of Biotechnology, National Dong Hwa University, Hualien 97401, Taiwan
| | - Yu-Fang Hu
- Pharmaceutical
Drug Delivery Division, TTY Biopharm Company Limited, Taipei 11469, Taiwan
| |
Collapse
|
35
|
Porter MR, Lindahl SE, Lietzke A, Metzger EM, Wang Q, Henck E, Chen CH, Niu H, Zaleski JM. Metal-mediated diradical tuning for DNA replication arrest via template strand scission. Proc Natl Acad Sci U S A 2017; 114:E7405-E7414. [PMID: 28760964 PMCID: PMC5594643 DOI: 10.1073/pnas.1621349114] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A series of M(PyED)·X (X = 2Cl-, SO42-) pyridine-metalloenediyne complexes [M = Cu(II), Fe(II), or Zn(II)] and their independently synthesized, cyclized analogs have been prepared to investigate their potential as radical-generating DNA-damaging agents. All complexes possess a 1:1 metal-to-ligand stoichiometry as determined by electronic absorption spectroscopy and X-ray diffraction. Solution structural analysis reveals a pπ Cl [Formula: see text] Cu(II) LMCT (22,026 cm-1) for Cu(PyED)·2Cl, indicating three nitrogens and a chloride in the psuedo-equatorial plane with the remaining pyridine nitrogen and solvent in axial positions. EPR spectra of the Cu(II) complexes exhibit an axially elongated octahedron. This spectroscopic evidence, together with density functional theory computed geometries, suggest six-coordinate structures for Cu(II) and Fe(II) complexes and a five-coordinate environment for Zn(II) analogs. Bergman cyclization via thermal activation of these constructs yields benzannulated product indicative of diradical generation in all complexes within 3 h at 37 °C. A significant metal dependence on the rate of the reaction is observed [Cu(II) > Fe(II) > Zn(II)], which is mirrored in in vitro DNA-damaging outcomes. Whereas in situ chelation of PyED leads to considerable degradation in the presence of all metals within 1 h under hyperthermia conditions, Cu(II) activation produces >50% compromised DNA within 5 min. Additionally, Cu(II) chelated PyED outcompetes DNA polymerase I to successfully inhibit template strand extension. Exposure of HeLa cells to Cu(PyBD)·SO4 (IC50 = 10 μM) results in a G2/M arrest compared with untreated samples, indicating significant DNA damage. These results demonstrate metal-controlled radical generation for degradation of biopolymers under physiologically relevant temperatures on short timescales.
Collapse
Affiliation(s)
- Meghan R Porter
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - Sarah E Lindahl
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - Anne Lietzke
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405
| | - Erin M Metzger
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - Quan Wang
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405
| | - Erik Henck
- Department of Chemistry, Indiana University, Bloomington, IN 47405
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405
| | - Chun-Hsing Chen
- Molecular Structure Center, Indiana University, Bloomington, IN 47405
| | - Hengyao Niu
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN 47405;
| | | |
Collapse
|
36
|
Fidler TP, Rowley JW, Araujo C, Boudreau LH, Marti A, Souvenir R, Dale K, Boilard E, Weyrich AS, Abel ED. Superoxide Dismutase 2 is dispensable for platelet function. Thromb Haemost 2017; 117:1859-1867. [PMID: 28771279 DOI: 10.1160/th17-03-0174] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 06/11/2017] [Indexed: 12/20/2022]
Abstract
Increased intracellular reactive oxygen species (ROS) promote platelet activation. The sources of platelet-derived ROS are diverse and whether or not mitochondrial derived ROS, modulates platelet function is incompletely understood. Studies of platelets from patients with sickle cell disease, and diabetes suggest a correlation between mitochondrial ROS and platelet dysfunction. Therefore, we generated mice with a platelet specific knockout of superoxide dismutase 2 (SOD2-KO) to determine if increased mitochondrial ROS increases platelet activation. SOD2-KO platelets demonstrated decreased SOD2 activity and increased mitochondrial ROS, however total platelet ROS was unchanged. Mitochondrial function and content were maintained in non-stimulated platelets. However SOD2-KO platelets demonstrated decreased mitochondrial function following thrombin stimulation. In vitro platelet activation and spreading was normal and in vivo, deletion of SOD2 did not change tail-bleeding or arterial thrombosis indices. In pathophysiological models mediated by platelet-dependent immune mechanisms such as sepsis and autoimmune inflammatory arthritis, SOD2-KO mice were phenotypically identical to wildtype controls. These data demonstrate that increased mitochondrial ROS does not result in platelet dysfunction.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - E Dale Abel
- E. Dale Abel, MB.BS., DPhil., Fraternal Order of Eagles Diabetes Research Center, Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, 4312 PBDB, 169 Newton Road, Iowa City, IA 52242-1101, USA, Tel.: +1 (319) 353 3050, Fax: +1 (319) 335 3865, E-mail:
| |
Collapse
|
37
|
Kosoko A, Olurinde O, Akinloye O. Doxorubicin induced neuro- and cardiotoxicities in experimental rats: Protection against oxidative damage by Theobroma cacao Stem bark. Biochem Biophys Rep 2017; 10:303-317. [PMID: 28955758 PMCID: PMC5614625 DOI: 10.1016/j.bbrep.2017.01.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 01/03/2017] [Accepted: 01/27/2017] [Indexed: 12/20/2022] Open
Abstract
80 rats, randomly selected, were divided into 3 treatment groups: pre-, co- and post-treatment; consisting of 6 sub-groups each (5 rats per sub-group): baseline, normal saline (2 mL), α-lipoic acid (20 mg/kg body weight), 200 mg/kg, 400 mg/kg or 800 mg/kg body weight Theobroma cacao stem bark aqueous extract (TCAE). All rats except for baseline group were intoxicated with 20 mg/kg body weight doxorubicin (DOX) intraperitoneally. The animals in pre- or post-treatment group received a single dose of DOX (20 mg/kg body weight) intraperitoneally 24 h before or after 7 days' oral administration with TCAE respectively while those in co-treatment group were co-administered 2.86 mg/kg body weight of DOX with either normal saline, α- lipoic acid or TCAE orally for 7 days. Animals were sacrificed (pre- and post- treatment groups were sacrificed on the ninth day while the co-treatment group sacrificed on the 8th day). Brain and heart tissue samples were harvested for enzyme markers of toxicity, oxidative stress and histopathological examinations. DOX intoxication caused significant decrease in activities of LDH and ACP, and increase in γGT and ALP activities in brain tissues while causing a significant increase in LDH, ACP, γGT activities and decrease in ALP activity in the cardiac tissues. DOX intoxication caused a significant increase in concentrations of H2O2 generated, MDA and PC, XO, MPx and NOX activities with concomitant decrease in CAT, SOD, GPx and GST activities, and in concentrations of GSH, AsA and α-Toc in brain and cardiac tissues. Pre-, co- and post-treatment with TCAE at either 200 mg/kg, 400 mg/kg or 800 mg/kg body weight significantly reversed the oxidative damage to the organs induced by DOX-intoxication. The result affirmed that T. cacao stem bark aqueous extract protected against DOX induced oxidative damage in brain and cardiac tissues of experimental rats.
Collapse
Affiliation(s)
- A.M. Kosoko
- Department of Biochemistry, College of Biosciences (COLBIOS), Federal University of Agriculture, Abeokuta (FUNAAB), Nigeria
| | | | | |
Collapse
|
38
|
Romanyuk AV, Grozdova ID, Ezhov AA, Melik-Nubarov NS. Peroxyoxalate Chemiluminescent Reaction as a Tool for Elimination of Tumour Cells Under Oxidative Stress. Sci Rep 2017; 7:3410. [PMID: 28611370 PMCID: PMC5469817 DOI: 10.1038/s41598-017-03527-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/28/2017] [Indexed: 12/13/2022] Open
Abstract
The overproduction of hydrogen peroxide is an inherent feature of some tumour cells and inflamed tissues. We took advantage of this peculiarity to eliminate cells using chemiluminescent peroxyoxalate reaction. We designed dispersions containing polyoxalate and tetramethylhematoporhyrin (TMHP) in dimethylphthalate droplets stabilized with Pluronic L64. The porphyrin plays the dual role. On the one hand, it serves as an activator of the peroxyoxalate reaction of polyoxalate with intracellular hydrogen peroxide and experiences excitation as a result of the reaction. The light emitted in the reaction in the model system without cells was used to optimize the dispersion's composition. On the other hand, TMHP acts as a photosensitizer (PS) causing cell damage. The formation of singlet oxygen led to cell elimination if the dispersions were used in combination with inducers of oxidative stress: hydrogen peroxide, paraquat, antitumour drug doxorubicin, or a nutritional additive menadione. The PS-induced cytotoxicity correlated with the level of intracellular ROS. The developed approach targeted to endogenous ROS is orthogonal to the classical chemotherapy and can be applied to increase its efficiency.
Collapse
Affiliation(s)
- Andrey V Romanyuk
- M.V. Lomonosov Moscow State University, Department of Chemistry, GSP-1, Leninskie gory 1, build. 3, Moscow, 119991, Russia
| | - Irina D Grozdova
- M.V. Lomonosov Moscow State University, Department of Chemistry, GSP-1, Leninskie gory 1, build. 3, Moscow, 119991, Russia
| | - Alexander A Ezhov
- M.V. Lomonosov Moscow State University, Department of Physics, GSP-1, Leninskie gory 1, build. 2, Moscow, 119991, Russia
| | - Nickolay S Melik-Nubarov
- M.V. Lomonosov Moscow State University, Department of Chemistry, GSP-1, Leninskie gory 1, build. 3, Moscow, 119991, Russia.
| |
Collapse
|
39
|
Quantitative determination of trace hydrogen peroxide in the presence of sulfide using the Amplex Red/horseradish peroxidase assay. Anal Chim Acta 2017; 963:61-67. [DOI: 10.1016/j.aca.2017.02.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 02/08/2017] [Accepted: 02/24/2017] [Indexed: 01/07/2023]
|
40
|
Kankala RK, Tsai PY, Kuthati Y, Wei PR, Liu CL, Lee CH. Overcoming multidrug resistance through co-delivery of ROS-generating nano-machinery in cancer therapeutics. J Mater Chem B 2017; 5:1507-1517. [DOI: 10.1039/c6tb03146c] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The use of nanotechnology to overcome multidrug resistance (MDR) in cancer cells has been predominant.
Collapse
Affiliation(s)
- Ranjith Kumar Kankala
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
- College of Chemical Engineering
| | - Pei-Yu Tsai
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
| | - Yaswanth Kuthati
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
| | - Pei-Ru Wei
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
| | - Chen-Lun Liu
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
| | - Chia-Hung Lee
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
| |
Collapse
|
41
|
Tong Q, Zhu Y, Galaske JW, Kosmacek EA, Chatterjee A, Dickinson BC, Oberley-Deegan RE. MnTE-2-PyP modulates thiol oxidation in a hydrogen peroxide-mediated manner in a human prostate cancer cell. Free Radic Biol Med 2016; 101:32-43. [PMID: 27671770 PMCID: PMC5486925 DOI: 10.1016/j.freeradbiomed.2016.09.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/20/2016] [Accepted: 09/23/2016] [Indexed: 01/11/2023]
Abstract
To improve the treatment of advanced prostate cancer, the development of effective and innovative antitumor agents is needed. Our previous work demonstrated that the ROS (reactive oxygen species) scavenger, MnTE-2-PyP, inhibited human prostate cancer growth and also inhibited prostate cancer migration and invasion. We showed that MnTE-2-PyP treatment altered the affinity of the histone acetyltransferase enzyme, p300, to bind to DNA. We speculate that this may be one mechanism by which MnTE-2-PyP inhibits prostate cancer progression. Specifically, MnTE-2-PyP decreased p300/HIF-1/CREB complex (p300/hypoxia-inducible factor-1/cAMP response element-binding protein) binding to a specific hypoxia-response element (HRE) motif within the plasminogen activator inhibitor-1 (PAI-1) gene promoter region, and consequently, repressed PAI-1 expression. However, it remains unclear how MnTE-2-PyP reduces p300 complex binding affinity to the promoter region of specific genes. In this study, we found that overexpression of Cu/ZnSOD (superoxide dismutase 1, SOD1) significantly suppressed PAI-1 gene expression and p300 complex binding to the promoter region of PAI-1 gene, just as was observed in cells treated with MnTE-2-PyP. Furthermore, catalase (CAT) overexpression rescued the inhibition of PAI-1 expression and p300 binding by MnTE-2-PyP. Taken together, the above findings suggest that hydrogen peroxide (H2O2) is likely the mediator through which MnTE-2-PyP inhibits the PAI-1 expression and p300 complex binding in PC3 cells. To confirm this, we measured the production of H2O2 following overexpression of SOD1 or catalase with MnTE-2-PyP treatment in the presence or absence of radiation. We found that MnTE-2-PyP increased the intracellular steady-state levels of H2O2 and increased nuclear H2O2 levels. As expected, catalase overexpression significantly decreased the levels of intracellular H2O2 induced by MnTE-2-PyP. We then determined if this increased H2O2 production could result in oxidized protein thiol groups. In the presence of MnTE-2-PyP, there was a significant increase in oxidized thiols in PC3 cell lysates and this was reversed with catalase overexpression. Specifically, we showed that p300 was oxidized after MnTE-2-PyP treatment, indicating that MnTE-2-PyP is creating a more oxidizing environment and this is altering the oxidation state of p300 thiol residues. Our data provide an in depth mechanism by which MnTE-2-PyP regulates gene transcription through induced H2O2 mediated oxidation of particular proteins, supporting an important role for MnTE-2-PyP as an effective and innovative antitumor agent to enhance treatment outcomes in prostate cancer radiotherapy.
Collapse
Affiliation(s)
- Qiang Tong
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuxiang Zhu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Joseph W Galaske
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Arpita Chatterjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bryan C Dickinson
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
42
|
Setyawati MI, Kutty RV, Leong DT. DNA Nanostructures Carrying Stoichiometrically Definable Antibodies. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2016; 12:5601-5611. [PMID: 27571230 DOI: 10.1002/smll.201601669] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Indexed: 05/08/2023]
Abstract
Targeted drug delivery is one of the key challenges in cancer nanomedicine. Stoichiometric and spatial control over the antibodies placement on the nanomedicine vehicle holds a pivotal role to overcome this key challenge. Here, a DNA tetrahedral is designed with available conjugation sites on its vertices, allowing to bind one, two, or three cetuximab antibodies per DNA nanostructure. This stoichiometrically definable cetuximab conjugated DNA nanostructure shows enhanced targeting on the breast cancer cells, which results with higher overall killing efficacy of the cancer cells.
Collapse
Affiliation(s)
- Magdiel Inggrid Setyawati
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Rajaletchumy Veloo Kutty
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
- Faculty of Chemical and Natural Resources Engineering, University Malaysia Pahang, Tun Razak Highway, 26300, Kuantan, Pahang, Malaysia
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore.
| |
Collapse
|
43
|
Lin B, McGuire K, Liu B, Jamison J, Tsai CC. Synthesis and anticancer activity of a hydroxytolan series. Bioorg Med Chem Lett 2016; 26:4451-4454. [PMID: 27515319 DOI: 10.1016/j.bmcl.2016.07.082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/29/2016] [Accepted: 07/30/2016] [Indexed: 11/28/2022]
Abstract
This paper describes the development of novel anticancer poly-hydroxylated tolans. Based on structural similarity to resveratrol, a series of hydroxytolans were synthesized and evaluated for their antitumor capability against three tumor cell lines and one fibroblast cell line for selectivity comparisons. The 4,4'-dihydroxytolan (KST-201) exhibited the most significant anticancer activity with increased selectivity when compared to resveratrol and other hydroxytolans. Unlike resveratrol, KST-201 can boost hydrogen peroxide in tumor cells, which are often at high basal level of reactive oxygen species, to cause cell death by overwhelming the cellular tolerance of oxidative stress.
Collapse
Affiliation(s)
- Boren Lin
- Department of Chemistry and Biochemistry, Kent State University, PO Box 5190, Kent, OH 44242-0001, United States
| | - Karen McGuire
- Department of Chemistry and Biochemistry, Kent State University, PO Box 5190, Kent, OH 44242-0001, United States
| | - Bo Liu
- Department of Chemistry and Biochemistry, Kent State University, PO Box 5190, Kent, OH 44242-0001, United States
| | - James Jamison
- Department of Urology, St. Thomas Hospital, Akron, OH 44310, United States
| | - Chun-Che Tsai
- Department of Chemistry and Biochemistry, Kent State University, PO Box 5190, Kent, OH 44242-0001, United States.
| |
Collapse
|
44
|
Ludwig JM, Gai Y, Sun L, Xiang G, Zeng D, Kim HS. SW43-DOX ± loading onto drug-eluting bead, a potential new targeted drug delivery platform for systemic and locoregional cancer treatment - An in vitro evaluation. Mol Oncol 2016; 10:1133-45. [PMID: 27262893 DOI: 10.1016/j.molonc.2016.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/19/2016] [Accepted: 05/12/2016] [Indexed: 12/31/2022] Open
Abstract
Treatment of unresectable primary cancer and their distant metastases, with the liver representing one of the most frequent location, is still plagued by insufficient treatment success and poor survival rates. The Sigma-2 receptor is preferentially expressed on many tumor cells making it an appealing target for therapy. Thus, we developed a potential targeted drug conjugate consisting of the Sigma-2 receptor ligand SW43 and Doxorubicin (SW43-DOX) for systemic cancer therapy and for locoregional treatment of primary and secondary liver malignancies when loaded onto drug-eluting bead (DEB) which was compared in vitro to the treatment with Doxorubicin alone. SW43-DOX binds specifically to the Sigma-2 receptor expressed on hepatocellular (Hep G2, Hep 3B), pancreatic (Panc-1) and colorectal (HT-29) carcinoma cell lines with high affinity and subsequent early specific internalization. Free SW43-DOX showed superior concentration and time depended cancer toxicity than treatment with Doxorubicin alone. Action mechanisms analysis revealed an apoptotic cell death with increased caspase 3/7 activation and reactive oxygen species (ROS) production. Only ROS scavenging with α-Tocopherol, but not the caspase inhibition (Z-VAD-FMK), partly reverted the effect. SW43-DOX could successfully be loaded onto DEB and showed prolonged eluting kinetics compared to Doxorubicin. SW43-DOX loaded DEB vs. Doxorubicin loaded DEB showed a significantly greater time dependent toxicity in all cell lines. In conclusion, the novel conjugate SW43-DOX ± loading onto DEB is a promising drug delivery platform for targeted systemic and locoregional cancer therapy.
Collapse
Affiliation(s)
- Johannes M Ludwig
- Division of Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 330 Cedar Street, New Haven, CT 06510, USA; Interventional Oncology Translational Laboratory, University of Pittsburgh School of Medicine, Presbyterian South Tower, 200 Lothrop Street, Pittsburgh, PA 15213-3553, USA
| | - Yongkang Gai
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh School of Medicine, 100 Technology Drive, Pittsburgh, PA 15219, USA
| | - Lingyi Sun
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh School of Medicine, 100 Technology Drive, Pittsburgh, PA 15219, USA
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China
| | - Dexing Zeng
- Molecular Imaging Laboratory, Department of Radiology, University of Pittsburgh School of Medicine, 100 Technology Drive, Pittsburgh, PA 15219, USA.
| | - Hyun S Kim
- Division of Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale School of Medicine, 330 Cedar Street, New Haven, CT 06510, USA; Interventional Oncology Translational Laboratory, University of Pittsburgh School of Medicine, Presbyterian South Tower, 200 Lothrop Street, Pittsburgh, PA 15213-3553, USA; Yale Cancer Center, Yale School of Medicine, New Haven, 330 Cedar Street, New Haven, CT 06510, USA.
| |
Collapse
|
45
|
Mukherjee AK, Saviola AJ, Burns PD, Mackessy SP. Apoptosis induction in human breast cancer (MCF-7) cells by a novel venom L-amino acid oxidase (Rusvinoxidase) is independent of its enzymatic activity and is accompanied by caspase-7 activation and reactive oxygen species production. Apoptosis 2015; 20:1358-72. [PMID: 26319994 DOI: 10.1007/s10495-015-1157-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
We report the elucidation of a mechanism of apoptosis induction in breast cancer (MCF-7) cells by an L-amino acid oxidase (LAAO), Rusvinoxidase, purified from the venom of Daboia russelii russelii. Peptide mass fingerprinting analysis of Rusvinoxidase, an acidic monomeric glycoprotein with a mass of ~57 kDa, confirmed its identity as snake venom LAAO. The enzymatic activity of Rusvinoxidase was completely abolished after two cycles of freezing and thawing; however, its cytotoxicity toward MCF-7 cells remained unaffected. Dose- and time-dependent induction of apoptosis by Rusvinoxidase on MCF-7 cells was evident from changes in cell morphology, cell membrane integrity, shrinkage of cells and apoptotic body formation accompanied by DNA fragmentation. Rusvinoxidase induced apoptosis in MCF-7 cells by both the extrinsic (death-receptor) and intrinsic (mitochondrial) signaling pathways. The former pathway of apoptosis operated through activation of caspase-8 that subsequently activated caspase-7 but not caspase-3. Rusvinoxidase-induced intrinsic pathway of apoptosis was accompanied by a time-dependent depolarization of the mitochondrial membrane through the generation of reactive oxygen species, followed by a decrease in cellular glutathione content and catalase activity, and down-regulation of expression of anti-apoptotic proteins Bcl-XL and heat-shock proteins (HSP-90 and HSP-70). Rusvinoxidase treatment resulted in increase of the pro-apoptotic protein Bax, subsequently leading to the release of cytochrome c from mitochondria to the cytosol and activating caspase-9, which in turn stimulated effector caspase-7. Rusvinoxidase at a dose of 4 mg/kg was non-toxic in mice, indicating that it may be useful as a model for the development of peptide-based anticancer drugs.
Collapse
Affiliation(s)
- Ashis K Mukherjee
- School of Biological Sciences, University of Northern Colorado, Greeley, CO, 80639-0017, USA,
| | | | | | | |
Collapse
|
46
|
Popovics P, Schally AV, Szalontay L, Block NL, Rick FG. Targeted cytotoxic analog of luteinizing hormone-releasing hormone (LHRH), AEZS-108 (AN-152), inhibits the growth of DU-145 human castration-resistant prostate cancer in vivo and in vitro through elevating p21 and ROS levels. Oncotarget 2015; 5:4567-78. [PMID: 24994120 PMCID: PMC4147346 DOI: 10.18632/oncotarget.2146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Management of castration-resistant prostate cancer (CRPC) is challenging due to lack of efficacious therapy. Luteinizing hormone-releasing hormone (LHRH) analogs appear to act directly on cells based on the LHRH receptors on human prostate adenocarcinoma cells. We explored anticancer activity of a cytotoxic analog of LHRH, AEZS-108, consisting of LHRH agonist linked to doxorubicin. Nude mice bearing DU-145 tumors were used to compare antitumor effects of AEZS-108 with its individual constituents or their unconjugated combination. The tumor growth inhibition of conjugate was greatest among treatment groups (90.5% inhibition vs. 41% by [D-Lys(6)]LHRH+DOX). The presence of LHRH receptors on DU-145 cells was confirmed by immunocytochemistry. In vitro, AEZS-108 significantly inhibited cell proliferation (61.2% inhibition) and elevated apoptosis rates (by 46%). By the detection of the inherent doxorubicin fluorescence, unconjugated doxorubicin was seen in the nucleus; the conjugate was perinuclear and at cell membrane. Autophagy, visualized by GFP-tagged p62 reporter, was increased by AEZS-108 (7.9-fold vs. 5.3-fold by DOX+[D-Lys(6)]LHRH. AEZS-108 more effectively increased reactive oxygen species (ROS, 2-fold vs. 1.4-fold by DOX+[D-Lys(6)]LHRH) and levels of the apoptotic regulator p21 in vivo and in vitro. We demonstrate robust inhibitory effects of the targeted cytotoxic LHRH analog, AEZS-108, on LHRHR positive castration-resistant prostate cancer cells.
Collapse
Affiliation(s)
- Petra Popovics
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Cardiovascular Diseases, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL; Department of Medicine III, Medical Faculty Carl Gustav Carus, Dresden, Germany
| | - Andrew V Schally
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL; Divisions of Hematology/Oncology, University of Miami, Miller School of Medicine, Miami, FL; Endocrinology University of Miami, Miller School of Medicine, Miami, FL
| | - Luca Szalontay
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL
| | - Norman L Block
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL; Divisions of Hematology/Oncology, University of Miami, Miller School of Medicine, Miami, FL
| | - Ferenc G Rick
- Veterans Affairs Medical Center and South Florida Veterans Affairs Foundation for Research and Education, Miami, FL; Department of Urology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL
| |
Collapse
|
47
|
Kankala RK, Kuthati Y, Liu CL, Mou CY, Lee CH. Killing cancer cells by delivering a nanoreactor for inhibition of catalase and catalytically enhancing intracellular levels of ROS. RSC Adv 2015. [DOI: 10.1039/c5ra16023e] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Intracellular hydrogen peroxide levels have the potential to be exploited in cancer therapy.
Collapse
Affiliation(s)
- Ranjith Kumar Kankala
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
| | - Yaswanth Kuthati
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
| | - Chen-Lun Liu
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
| | - Chung-Yuan Mou
- Department of Chemistry
- National Taiwan University
- Taipei 106
- Taiwan
| | - Chia-Hung Lee
- Department of Life Science and Institute of Biotechnology
- National Dong Hwa University
- Hualien
- Taiwan
| |
Collapse
|
48
|
Luo C, Li Y, Zhou B, Yang L, Li H, Feng Z, Li Y, Long J, Liu J. A monocarbonyl analogue of curcumin, 1,5-bis(3-hydroxyphenyl)-1,4-pentadiene-3-one (Ca 37), exhibits potent growth suppressive activity and enhances the inhibitory effect of curcumin on human prostate cancer cells. Apoptosis 2014; 19:542-53. [PMID: 24297639 DOI: 10.1007/s10495-013-0947-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Prostate carcinoma is one of the leading causes of cancer-related morbidity and mortality in males in western countries. Curcumin exhibits growth-suppressive activity against several cancers, including prostate cancer, but it has poor bioavailability. The purpose of this study was to evaluate the anticancer potency and mechanism of a curcumin analogue, 1,5-bis(3-hydroxyphenyl)-1,4-pentadiene-3-one (Ca 37), in human prostate cancer. Studies were performed in established human prostate cancer cell lines (PC-3 and DU145) as well as in a murine xenograft tumor (PC-3) model. Ca 37 presented a preferential suppression capacity against growth and migration toward prostate cancer cells compared with curcumin. Ca 37 impaired the bioenergetics system, promoted cell cycle arrest and apoptosis activation in PC-3 cells. In addition, 0.5 μmol (6.65 mg/kg body weight) of Ca 37 significantly inhibited the growth of the prostate xenografted tumors, whereas 6 μmol (110 mg/kg body weight) of curcumin had little effect. Furthermore, a combination of Ca 37 and curcumin resulted in enhanced antitumor activity in prostate cancer cells. N-Acetylcysteine abrogated both reactive oxygen species (ROS) production and viability loss induced by Ca 37 but partially prevented growth inhibition in PC-3 cells treated with curcumin alone, or a combination with Ca 37. The data indicate that induction of ROS plays a vital role in the growth inhibitory effect of Ca 37 in PC-3 cells. This study suggests that Ca 37, alone or in combination with curcumin, may be a promising anticancer agent for prostate cancer therapy.
Collapse
Affiliation(s)
- Cheng Luo
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology and Frontier Institute of Life Science, FIST, Xi'an Jiaotong University, Xi'an, 710049, China,
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Kanugula AK, Gollavilli PN, Vasamsetti SB, Karnewar S, Gopoju R, Ummanni R, Kotamraju S. Statin‐induced inhibition of breast cancer proliferation and invasion involves attenuation of iron transport: intermediacy of nitric oxide and antioxidant defence mechanisms. FEBS J 2014; 281:3719-3738. [DOI: https:/doi.org/10.1111/febs.12893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 06/23/2014] [Indexed: 11/07/2023]
Abstract
Accumulating evidence from in vitro, in vivo, clinical and epidemiological studies shows promising results for the use of statins against many cancers including breast carcinoma. However, the molecular mechanisms responsible for the anti‐proliferative and anti‐invasive properties of statins still remain elusive. In this study, we investigated the involvement of nitric oxide, iron homeostasis and antioxidant defence mechanisms in mediating the anti‐proliferative and anti‐invasive properties of hydrophobic statins in MDA‐MB‐231, MDA‐MB‐453 and BT‐549 metastatic triple negative breast cancer cells. Fluvastatin and simvastatin significantly increased cytotoxicity which was reversed with mevalonate. Interestingly, fluvastatin downregulated transferrin receptor (TfR1), with a concomitant depletion of intracellular iron levels in these cells. Statin‐induced effects were mimicked by geranylgeranyl transferase inhibitor (GGTI‐298) but not farnesyl transferase inhibitor (FTI‐277). Further, it was observed that TfR1 downregulation is mediated by increased nitric oxide levels via inducible nitric oxide synthase (iNOS) expression. NOS inhibitors (asymmetric dimethylarginine and 1400W) counteracted and sepiapterin, a precursor of tetrahydrobiopterin, exacerbated statin‐induced depletion of intracellular iron levels. Notably, fluvastatin increased manganese superoxide dismutase (by repressing the transcription factor DNA damage‐binding protein 2), catalase and glutathione which, in turn, diminished H2O2 levels. Fluvastatin‐induced downregulation of TfR1, matrix metalloproteinase‐2, ‐9 and inhibition of invasion were reversed in the presence of aminotriazole, a specific inhibitor of catalase. Finally, we conclude that fluvastatin, by altering iron homeostasis, nitric oxide generation and antioxidant defence mechanisms, induces triple negative breast cancer cell death.
Collapse
Affiliation(s)
| | | | | | - Santosh Karnewar
- Centre for Chemical Biology CSIR Indian Institute of Chemical Technology Hyderabad India
| | - Raja Gopoju
- Centre for Chemical Biology CSIR Indian Institute of Chemical Technology Hyderabad India
| | - Ramesh Ummanni
- Centre for Chemical Biology CSIR Indian Institute of Chemical Technology Hyderabad India
| | - Srigiridhar Kotamraju
- Centre for Chemical Biology CSIR Indian Institute of Chemical Technology Hyderabad India
| |
Collapse
|
50
|
Kanugula AK, Gollavilli PN, Vasamsetti SB, Karnewar S, Gopoju R, Ummanni R, Kotamraju S. Statin-induced inhibition of breast cancer proliferation and invasion involves attenuation of iron transport: intermediacy of nitric oxide and antioxidant defence mechanisms. FEBS J 2014; 281:3719-38. [PMID: 24964743 DOI: 10.1111/febs.12893] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 06/05/2014] [Accepted: 06/23/2014] [Indexed: 11/29/2022]
Abstract
Accumulating evidence from in vitro, in vivo, clinical and epidemiological studies shows promising results for the use of statins against many cancers including breast carcinoma. However, the molecular mechanisms responsible for the anti-proliferative and anti-invasive properties of statins still remain elusive. In this study, we investigated the involvement of nitric oxide, iron homeostasis and antioxidant defence mechanisms in mediating the anti-proliferative and anti-invasive properties of hydrophobic statins in MDA-MB-231, MDA-MB-453 and BT-549 metastatic triple negative breast cancer cells. Fluvastatin and simvastatin significantly increased cytotoxicity which was reversed with mevalonate. Interestingly, fluvastatin downregulated transferrin receptor (TfR1), with a concomitant depletion of intracellular iron levels in these cells. Statin-induced effects were mimicked by geranylgeranyl transferase inhibitor (GGTI-298) but not farnesyl transferase inhibitor (FTI-277). Further, it was observed that TfR1 downregulation is mediated by increased nitric oxide levels via inducible nitric oxide synthase (iNOS) expression. NOS inhibitors (asymmetric dimethylarginine and 1400W) counteracted and sepiapterin, a precursor of tetrahydrobiopterin, exacerbated statin-induced depletion of intracellular iron levels. Notably, fluvastatin increased manganese superoxide dismutase (by repressing the transcription factor DNA damage-binding protein 2), catalase and glutathione which, in turn, diminished H2 O2 levels. Fluvastatin-induced downregulation of TfR1, matrix metalloproteinase-2, -9 and inhibition of invasion were reversed in the presence of aminotriazole, a specific inhibitor of catalase. Finally, we conclude that fluvastatin, by altering iron homeostasis, nitric oxide generation and antioxidant defence mechanisms, induces triple negative breast cancer cell death.
Collapse
|