1
|
Shokrzadeh S, Moghim S, Shokrzadeh M, Aghajanshakeri S. An added value of azithromycin: mitigation of doxorubicin-associated oxidative damage and genotoxicity in normal human bronchial epithelium cells. Mutagenesis 2025; 40:126-136. [PMID: 39373202 DOI: 10.1093/mutage/geae024] [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: 06/13/2024] [Accepted: 10/05/2024] [Indexed: 10/08/2024] Open
Abstract
Doxorubicin, a well-known and widely used antineoplastic agent with direct ROS-accumulating activity, has proven effective in treating various cancer types. However, its non-specific cytotoxicity towards non-cancerous cells prompts concerns regarding potential adverse effects. Azithromycin is an antibiotic for treating bacterial infections and an anti-inflammatory agent, particularly beneficial in managing respiratory conditions like bronchitis and sinusitis. Despite azithromycin's well-documented antibacterial properties, its potential cellular/genomic protective effects remain unexplored. As an in vitro model, BEAS-2B cells (normal human bronchial epithelium cells) were employed in this study to assess whether azithromycin possesses any protective properties against doxorubicin-induced cellular toxicity. Cells in pretreatment culture were treated to various amounts of azithromycin (3.125, 6.25, 12.5, 25, and 50 μg/ml) in combination with doxorubicin at IC50 (0.08 μg/ml). Doxorubicin at 0.08 μg/ml highlighted cytotoxicity, oxidative stress, and genotoxicity. Azithromycin at 25 and 50 μg/ml markedly modulated oxidative stress and genomic damage by decreasing the ROS and LPO amounts and suppressing DNA fragmentation in the comet assay parameters. Consequently, azithromycin may be regarded as a cytomodulating, antigenotoxic, and antioxidant agent.
Collapse
Affiliation(s)
| | - Shahrzad Moghim
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Shokrzadeh
- Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shaghayegh Aghajanshakeri
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
2
|
Uurasmaa TM, Bourdin P, Nammas W, Latifi S, Liljenbäck H, Saraste A, Eskola O, Rajander J, Roivainen A, Rundqvist H, Autio A, Heinonen I, Anttila K. Exercise training partly ameliorates cardiac dysfunction in mice during doxorubicin treatment of breast cancer. J Transl Med 2025; 23:89. [PMID: 39838445 PMCID: PMC11748283 DOI: 10.1186/s12967-025-06108-y] [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: 10/07/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
INTRODUCTION Doxorubicin is a chemotherapeutic drug used to treat various cancers. Exercise training (ET) can attenuate some cardiotoxic effects of doxorubicin (DOX) in tumor-free animals. However, the ET effects on cardiac function and glucose metabolism in DOX-treated breast cancer models remain unclear. OBJECTIVES This study investigated ET-induced structural, functional, vascular, oxidative stress, and plausible glucose uptake alterations of the left ventricle (LV) in a murine breast cancer model during DOX treatment. METHODS Female FVB/N-mice were divided to tumor-free groups with or without voluntary wheel-running ET and those inoculated subcutaneously with mammary tumor-derived I3TC-cells with or without exercise or DOX treatment (5 mg/kg/week). Mice underwent 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography and echocardiography after two and four DOX-doses. The cardiac histology, oxidative stress, maximal metabolic enzyme activities, and mitochondrial respiration were analyzed. RESULTS DOX increased LV glucose uptake (LVGU) and mitochondrial uncoupling and decreased running activity, LV-weight, and ejection fraction (EF). In DOX-treated group ET blunted the increase in LVGU, increased LV-weight and EF, and lowered LV lactate dehydrogenase activity. DOX-treated exercised mice did not differ from tumor-bearing group without DOX in LVGU or from the tumor-free ET-group in LV-weight or EF whereas unexercised DOX-treated group did. ET also increased LV citrate synthase activity in tumor-bearing animals. There was an inverse association between LVGU and EF and LV-weight. CONCLUSION In a murine breast cancer model, voluntary ET moderated DOX-induced cardiotoxicities such as increased LVGU, LV-atrophy and decreased EF. This suggests that ET might benefit patients with cancer undergoing doxorubicin treatment by mitigating cardiotoxicity.
Collapse
Affiliation(s)
- Tytti-Maria Uurasmaa
- Department of Biology, University of Turku, Turku, Finland.
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland.
| | | | - Wail Nammas
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
- Heart Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Shiva Latifi
- Department of Biology, University of Turku, Turku, Finland
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
- Heart Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Olli Eskola
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Johan Rajander
- Accelerator Laboratory, Åbo Akademi University, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Helene Rundqvist
- Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anu Autio
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Ilkka Heinonen
- Turku PET Centre, University of Turku, Turku University Hospital, Turku, Finland
| | - Katja Anttila
- Department of Biology, University of Turku, Turku, Finland
| |
Collapse
|
3
|
Bisht A, Avinash D, Sahu KK, Patel P, Das Gupta G, Kurmi BD. A comprehensive review on doxorubicin: mechanisms, toxicity, clinical trials, combination therapies and nanoformulations in breast cancer. Drug Deliv Transl Res 2025; 15:102-133. [PMID: 38884850 DOI: 10.1007/s13346-024-01648-0] [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] [Accepted: 05/31/2024] [Indexed: 06/18/2024]
Abstract
Doxorubicin is a key treatment for breast cancer, but its effectiveness often comes with significant side effects. Its actions include DNA intercalation, topoisomerase II inhibition, and reactive oxygen species generation, leading to DNA damage and cell death. However, it can also cause heart problems and low blood cell counts. Current trials aim to improve doxorubicin therapy by adjusting doses, using different administration methods, and combining it with targeted treatments or immunotherapy. Nanoformulations show promise in enhancing doxorubicin's effectiveness by improving drug delivery, reducing side effects, and overcoming drug resistance. This review summarizes recent progress and difficulties in using doxorubicin for breast cancer, highlighting its mechanisms, side effects, ongoing trials, and the potential impact of nanoformulations. Understanding these different aspects is crucial in optimizing doxorubicin's use and improving outcomes for breast cancer patients. This review examines the toxicity of doxorubicin, a drug used in breast cancer treatment, and discusses strategies to mitigate adverse effects, such as cardioprotective agents and liposomal formulations. It also discusses clinical trials evaluating doxorubicin-based regimens, the evolving landscape of combination therapies, and the potential of nanoformulations to optimize delivery and reduce systemic toxicity. The review also discusses the potential of liposomes, nanoparticles, and polymeric micelles to enhance drug accumulation within tumor tissues while sparing healthy organs.
Collapse
Affiliation(s)
- Anjali Bisht
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Dubey Avinash
- Department of Pharmaceutical Quality Assurance, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, 17 km Stone, NH-2, Chaumuhan, Mathura, 281406, UP, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
4
|
Selyutina OY, Ul’yanova MA, Chinak OA, Timoshnikov VA, Fedenok LG, Stepanov AA, Yanshole VV, Kulik LV, Vasilevsky SF, Polyakov NE, Kontoghiorghes GJ. Novel Anthraquinone Derivatives and Their Complexes with Metal Ions with Anticancer Activity: Structure/Redox and Chelation Activity Correlations. Pharmaceuticals (Basel) 2024; 17:1717. [PMID: 39770559 PMCID: PMC11678833 DOI: 10.3390/ph17121717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Some specific anthraquinone derivatives (AQs) are known to be used widely as effective chemotherapeutic agents in the treatment of cancer. However, their fundamental shortcoming is the high rate of cardiotoxicity observed in treated patients, which is thought to be caused by the increase in production of reactive oxygen species (ROS) catalyzed by iron and copper. The development of improved AQs and other anticancer drugs with enhanced efficacy but reduced toxicity remains a high priority. The aim of this study was to evaluate the cytotoxic and ROS production effects of chelate iron and copper complexes of two novel AQs, namely 4-hydroxynaphto[2,3-h]cinnoline-7,12-dione (Q2) and 3-(hydroxymethyl)naphto[2,3-h]cinnoline-4,7,12(1H)-trione (Q3). Methods: The chelation ability of Q2 and Q3 was studied using NMR and UV-Vis spectroscopy. Cytotoxicity studies were carried out using the MTT assay. The influence of chelation on ROS production was studied using NMR spectroscopy in linoleic acid micelles. Results: It was found that only Q3 forms complexes with Fe(III) and Cu(II) ions, whereas Q2 does not demonstrate chelating properties. A cytotoxicity study revealed that Fe[Q3]3 significantly decreased the viability of lung cancer A549 cells, while Q3 and Cu[Q3]2 did not demonstrate cytotoxic properties in this cell line. Furthermore, the presence of Q3 lowered the rate of iron-induced lipid peroxidation in linoleic acid micelles. By contrast, Q2 did not influence the rate of lipid peroxidation, probably due to the absence of effective metal chelating ability. Conclusions: The high cytotoxic effects observed with the iron complex of Q3 against cancer cells in combination with a reduced rate of iron induced lipid peroxidation in the presence of Q3, make Q3 and its iron complex promising for further evaluation and use as chemotherapeutic agents in cancer.
Collapse
Affiliation(s)
- Olga Yu. Selyutina
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Maya A. Ul’yanova
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Olga A. Chinak
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Lavrentyev Ave. 8, Novosibirsk 630090, Russia
| | - Viktor A. Timoshnikov
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Lidiya G. Fedenok
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Alexander A. Stepanov
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Vadim V. Yanshole
- International Tomography Center SB RAS, Institutskaya Str. 3a, Novosibirsk 630090, Russia;
| | - Leonid V. Kulik
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Sergey F. Vasilevsky
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - Nikolay E. Polyakov
- Institute of Chemical Kinetics and Combustion, Institutskaya Str. 3, Novosibirsk 630090, Russia; (O.Y.S.); (M.A.U.); (O.A.C.); (V.A.T.); (L.G.F.); (A.A.S.); (L.V.K.); (N.E.P.)
| | - George J. Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol CY-3021, Cyprus
| |
Collapse
|
5
|
Stachowicz-Kuśnierz A, Rychlik P, Korchowiec J, Korchowiec B. Insights into Transfer of Supramolecular Doxorubicin/Congo Red Aggregates through Phospholipid Membranes. Molecules 2024; 29:2567. [PMID: 38893446 PMCID: PMC11173802 DOI: 10.3390/molecules29112567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Doxorubicin (DOX) is a commonly used chemotherapeutic drug, from the anthracycline class, which is genotoxic to neoplastic cells via a DNA intercalation mechanism. It is effective and universal; however, it also causes numerous side effects. The most serious of them are cardiotoxicity and a decrease in the number of myeloid cells. For this reason, targeted DOX delivery systems are desirable, since they would allow lowering the drug dose and therefore limiting systemic side effects. Recently, synthetic dyes, in particular Congo red (CR), have been proposed as possible DOX carriers. CR is a planar molecule, built of a central biphenyl moiety and two substituted naphthalene rings, connected with diazo bonds. In water, it forms elongated ribbon-shaped supramolecular structures, which are able to selectively interact with immune complexes. In our previous studies, we have shown that CR aggregates can intercalate DOX molecules. In this way, they preclude DOX precipitation in water solutions and increase its uptake by MCF7 breast cancer cells. In the present work, we further explore the interactions between DOX, CR, and their aggregates (CR/DOX) with phospholipid membranes. In addition to neutral molecules, the protonated doxorubicin form, DXP, is also studied. Molecular dynamics simulations are employed to study the transfer of CR, DOX, DXP, and their aggregates through POPC bilayers. Interactions of CR, DOX, and CR/DOX with model monolayers are studied with Langmuir trough measurements. This study shows that CR may support the transfer of doxorubicin molecules into the bilayer. Both electrostatic and van der Waals interactions with lipids are important in this respect. The former promote the initial stages of the insertion process, the latter keep guest molecules inside the bilayer.
Collapse
Affiliation(s)
- Anna Stachowicz-Kuśnierz
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; (P.R.); (J.K.)
| | | | | | - Beata Korchowiec
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland; (P.R.); (J.K.)
| |
Collapse
|
6
|
Selyutina OY, Babenko SV, Slepneva IA, Polyakov NE, Kontoghiorghes GJ. Increased Free Radical Generation during the Interaction of a Quinone-Quinoline Chelator with Metal Ions and the Enhancing Effect of Light. Pharmaceuticals (Basel) 2023; 16:1116. [PMID: 37631031 PMCID: PMC10459951 DOI: 10.3390/ph16081116] [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: 07/04/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Schiff bases and similar molecules forming metal complexes may cause redox effects, which may also be influenced by light. Anthraquinones such as doxorubicin and idarubicin are widely used antitumor agents, which can generate reactive oxygen species (ROS), stimulated by both the presence of iron and copper ions and also by light. The generated ROS can cause DNA scission, cell membrane oxidation, and many other toxic effects. The redox activity of the quinone-quinoline chelator 2-phenyl-4-(butylamino)naphtho [2,3-h]quinoline-7,12-dione (Q1) was investigated in the presence of iron, copper, and zinc. The influence of light in these interactions was also examined. The chemically induced dynamic nuclear polarization (CIDNP), nuclear magnetic resonance (NMR), and electron paramagnetic resonance (EPR) methods were used to elucidate the molecular changes and ROS generation effects of the Q1 metal interactions. A model electron transfer reaction system between 1,4-dihydropyridine and Q1 was utilized to demonstrate that the chelate complexes of Q1 with both Fe(III) and Cu(II) ions were more redox active than Q1 itself. Similarly, CIDNP and NMR data showed that the concentration dependence of the free radicals yield is much higher in the presence of Fe(III) and Cu(II) ions, in comparison to Zn(II), and also that it increased in the presence of light. These findings underline the role of transition metal ions and Q1 in cyclic redox chain reactions and increase the prospect of the development of copper- and iron-based chelating agents, including Q1 and its derivatives, for anticancer therapy. Furthermore, these findings also signify the effect of light on enhancing ROS formation by Q1 and the prospect of utilizing such information for designing target specific anticancer drugs for photodynamic therapy.
Collapse
Affiliation(s)
- Olga Yu. Selyutina
- Institute of Chemical Kinetics & Combustion, Novosibirsk 630090, Russia; (O.Y.S.); (S.V.B.); (I.A.S.); (N.E.P.)
| | - Simon V. Babenko
- Institute of Chemical Kinetics & Combustion, Novosibirsk 630090, Russia; (O.Y.S.); (S.V.B.); (I.A.S.); (N.E.P.)
- International Tomography Center, Novosibirsk 630090, Russia
| | - Irina A. Slepneva
- Institute of Chemical Kinetics & Combustion, Novosibirsk 630090, Russia; (O.Y.S.); (S.V.B.); (I.A.S.); (N.E.P.)
| | - Nikolay E. Polyakov
- Institute of Chemical Kinetics & Combustion, Novosibirsk 630090, Russia; (O.Y.S.); (S.V.B.); (I.A.S.); (N.E.P.)
| | - George J. Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, Limassol CY-3021, Cyprus
| |
Collapse
|
7
|
Sada M, Matsushima S, Ikeda M, Ikeda S, Okabe K, Ishikita A, Tadokoro T, Enzan N, Yamamoto T, Miyamoto HD, Tsutsui Y, Miyake R, Setoyama D, Kang D, Ide T, Tsutsui H. IFN-γ-STAT1-ERK Pathway Mediates Protective Effects of Invariant Natural Killer T Cells Against Doxorubicin-Induced Cardiomyocyte Death. JACC Basic Transl Sci 2023; 8:992-1007. [PMID: 37719427 PMCID: PMC10504401 DOI: 10.1016/j.jacbts.2023.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 09/19/2023]
Abstract
Doxorubicin (DOX)-induced cardiomyopathy has poor prognosis, and myocardial inflammation is intimately involved in its pathophysiology. The role of invariant natural killer T (iNKT) cells has not been fully determined in this disease. We here demonstrated that activation of iNKT cells by α-galactosylceramide (GC) attenuated DOX-induced cardiomyocyte death and cardiac dysfunction. αGC increased interferon (IFN)-γ and phosphorylation of signal transducers and activators of transcription 1 (STAT1) and extracellular signal-regulated kinase (ERK). Administration of anti-IFN-γ neutralizing antibody abrogated the beneficial effects of αGC on DOX-induced cardiac dysfunction. These findings emphasize the protective role of iNKT cells in DOX-induced cardiomyopathy via the IFN-γ-STAT1-ERK pathway.
Collapse
Affiliation(s)
- Masashi Sada
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Soichiro Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kosuke Okabe
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akihito Ishikita
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomonori Tadokoro
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuyuki Enzan
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taishi Yamamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroko Deguchi Miyamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshitomo Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Miyake
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dongchon Kang
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cardiovascular Medicine, Research Institute of Angiocardiology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
Becker MMC, Arruda GFA, Berenguer DRF, Buril RO, Cardinale D, Brandão SCS. Anthracycline cardiotoxicity: current methods of diagnosis and possible role of 18F-FDG PET/CT as a new biomarker. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:17. [PMID: 36973762 PMCID: PMC10041777 DOI: 10.1186/s40959-023-00161-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 02/01/2023] [Indexed: 03/29/2023]
Abstract
Despite advances in chemotherapy, the drugs used in cancer treatment remain rather harmful to the cardiovascular system, causing structural and functional cardiotoxic changes. Positron-emission tomography associated with computed tomography (PET/CT) has emerged like a promising technique in the early diagnosis of these adverse drug effects as the myocardial tissue uptake of fluorodeoxyglucose labeled with fluorine-18 (18F-FDG), a glucose analog, is increased after their use. Among these drugs, anthracyclines are the most frequently associated with cardiotoxicity because they promote heart damage through DNA breaks, and induction of an oxidative, proinflammatory, and toxic environment. This review aimed to present the scientific evidence available so far regarding the use of 18F-FDG PET/CT as an early biomarker of anthracycline-related cardiotoxicity. Thus, it discusses the physiological basis for its uptake, hypotheses to justify its increase in the myocardium affected by anthracyclines, importance of 18F-FDG PET/CT findings for cardio-oncology, and primary challenges of incorporating this technique in standard clinical oncology practice.
Collapse
Affiliation(s)
- Mônica M C Becker
- Postgraduate Program in Surgery, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Gustavo F A Arruda
- Recife Medical School, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Diego R F Berenguer
- Postgraduate Program in Translational Health, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Roberto O Buril
- Postgraduate Program in Surgery, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, I.R.C.C.S., Milan, Italy
| | - Simone C S Brandão
- Postgraduate Program in Surgery, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil.
- Recife Medical School, Federal University of Pernambuco, Recife, State of Pernambuco, Brazil.
- Nuclear Medicine Department, Hospital das Clínicas, Federal University of Pernambuco, 1st floor, 1235 Avenida Professor Moraes Rego, Recife, State of Pernambuco, 50670-901, Brazil.
| |
Collapse
|
9
|
Kciuk M, Gielecińska A, Mujwar S, Kołat D, Kałuzińska-Kołat Ż, Celik I, Kontek R. Doxorubicin-An Agent with Multiple Mechanisms of Anticancer Activity. Cells 2023; 12:659. [PMID: 36831326 PMCID: PMC9954613 DOI: 10.3390/cells12040659] [Citation(s) in RCA: 214] [Impact Index Per Article: 107.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Doxorubicin (DOX) constitutes the major constituent of anti-cancer treatment regimens currently in clinical use. However, the precise mechanisms of DOX's action are not fully understood. Emerging evidence points to the pleiotropic anticancer activity of DOX, including its contribution to DNA damage, reactive oxygen species (ROS) production, apoptosis, senescence, autophagy, ferroptosis, and pyroptosis induction, as well as its immunomodulatory role. This review aims to collect information on the anticancer mechanisms of DOX as well as its influence on anti-tumor immune response, providing a rationale behind the importance of DOX in modern cancer therapy.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Somdutt Mujwar
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Damian Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, 90-136 Lodz, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Experimental Surgery, Faculty of Medicine, Medical University of Lodz, 90-136 Lodz, Poland
| | - Ismail Celik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, 38039 Kayseri, Turkey
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland
| |
Collapse
|
10
|
Selyutina OY, Mastova AV, Polyakov NE. The Interaction of Anthracycline Based Quinone-Chelators with Model Lipid Membranes: 1H NMR and MD Study. MEMBRANES 2023; 13:membranes13010061. [PMID: 36676868 PMCID: PMC9861344 DOI: 10.3390/membranes13010061] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 06/01/2023]
Abstract
Anthracycline antibiotics, e.g., doxorubicin, daunomycin, and other anthraquinones, are an important family of antitumor agents widely used in chemotherapy, which is currently the principal method for treating many malignancies. Thus, development of improved antitumor drugs with enhanced efficacy remains a high priority. Interaction of anthraquinone-based anticancer drugs with cell membranes attracts significant attention due to its importance in the eventual overcoming of multidrug resistance (MDR). The use of drugs able to accumulate in the cell membrane is one of the possible ways of overcoming MDR. In the present work, the aspects of interaction of anthraquinone 2-phenyl-4-(butylamino)naphtho[2,3-h]quinoline-7,12-dione) (Q1) with a model membrane were studied by means of NMR and molecular dynamics simulations. A fundamental shortcoming of anthracycline antibiotics is their high cardiotoxicity caused by reactive oxygen species (ROS). The important feature of Q1 is its ability to chelate transition metal ions responsible for ROS generation in vivo. In the present study, we have shown that Q1 and its chelating complexes penetrated into the lipid membrane and were located in the hydrophobic part of the bilayer near the bilayer surface. The chelate complex formation of Q1 with metal ions increased its penetration ability. In addition, it was found that the interaction of Q1 with lipid molecules could influence lipid mobility in the bilayer. The obtained results have an impact on the understanding of molecular mechanisms of Q1 biological activity.
Collapse
|
11
|
Giráldez-Pérez RM, Grueso E, Montero-Hidalgo AJ, Luque RM, Carnerero JM, Kuliszewska E, Prado-Gotor R. Gold Nanosystems Covered with Doxorubicin/DNA Complexes: A Therapeutic Target for Prostate and Liver Cancer. Int J Mol Sci 2022; 23:ijms232415575. [PMID: 36555216 PMCID: PMC9779246 DOI: 10.3390/ijms232415575] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Different gold nanosystems covered with DNA and doxorubicin (Doxo) were designed and synthesized for cancer therapy, starting from Au@16-Ph-16 cationic nanoparticles and DNA-Doxo complexes prepared under saturation conditions. For the preparation of stable, biocompatible, and small-sized compacted Au@16-Ph-16/DNA-Doxo nanotransporters, the conditions for the DNA-Doxo compaction process induced by gold nanoparticles were first explored using fluorescence spectroscopy, circular dichroism and atomic force microscopy techniques. The reverse process, which is fundamental for Doxo liberation at the site of action, was found to occur at higher CAu@16-Ph-16 concentrations using these techniques. Zeta potential, dynamic light scattering and UV-visible spectroscopy reveal that the prepared compacted nanosystems are stable, highly charged and of adequate size for the effective delivery of Doxo to the cell. This fact is verified by in vitro biocompatibility and internalization studies using two prostate cancer-derived cell lines (LNCaP and DU145) and one hepatocellular carcinoma-derived cell line (SNU-387), as well as a non-tumor prostate (PNT2) cell line and a non-hepatocarcinoma hepatoblastoma cell line (Hep-G2) model used as a control in liver cells. However, the most outstanding results of this work are derived from the use of the CI+NI combined treatments which present strong action in cancer-derived cell lines, while a protective effect is observed in non-tumor cell lines. Hence, novel therapeutic targets based on gold nanoparticles denote high selectivity compared to conventional treatment based on free Doxo at the same concentration. The results obtained show the viability of both the proposed methodology for internalization of compacted nanocomplexes inside the cell and the effectiveness of the possible treatment and minimization of side effects in prostate and liver cancer.
Collapse
Affiliation(s)
- Rosa M. Giráldez-Pérez
- Department of Cell Biology, Physiology and Immunology, Faculty of Sciences, University of Cordoba, 14014 Cordoba, Spain
- Correspondence: (R.M.G.-P.); (E.G.)
| | - Elia Grueso
- Department of Physical Chemistry, Faculty of Chemistry, University of Seville, 41012 Seville, Spain
- Correspondence: (R.M.G.-P.); (E.G.)
| | - Antonio J. Montero-Hidalgo
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital (HURS), Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Raúl M. Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Reina Sofia University Hospital (HURS), Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - José M. Carnerero
- Department of Physical Chemistry, Faculty of Chemistry, University of Seville, 41012 Seville, Spain
| | | | - Rafael Prado-Gotor
- Department of Physical Chemistry, Faculty of Chemistry, University of Seville, 41012 Seville, Spain
| |
Collapse
|
12
|
Ling G, Wang X, Tan N, Cao J, Li W, Zhang Y, Jiang J, Sun Q, Jiang Y, Wang W, Wang Y. Mechanisms and Drug Intervention for Doxorubicin-Induced Cardiotoxicity Based on Mitochondrial Bioenergetics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7176282. [PMID: 36275901 PMCID: PMC9586735 DOI: 10.1155/2022/7176282] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/17/2022] [Accepted: 09/10/2022] [Indexed: 11/18/2022]
Abstract
Doxorubicin (DOX) is an anthracycline chemotherapy drug, which is indispensable in antitumor therapy. However, its subsequent induction of cardiovascular disease (CVD) has become the primary cause of mortality in cancer survivors. Accumulating evidence has demonstrated that cardiac mitochondrial bioenergetics changes have become a significant marker for doxorubicin-induced cardiotoxicity (DIC). Here, we mainly summarize the related mechanisms of DOX-induced cardiac mitochondrial bioenergetics disorders reported in recent years, including mitochondrial substrate metabolism, the mitochondrial respiratory chain, myocardial ATP storage and utilization, and other mechanisms affecting mitochondrial bioenergetics. In addition, intervention for DOX-induced cardiac mitochondrial bioenergetics disorders using chemical drugs and traditional herbal medicine is also summarized, which will provide a comprehensive process to study and develop more appropriate therapeutic strategies for DIC.
Collapse
Affiliation(s)
- Guanjing Ling
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoping Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Nannan Tan
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jing Cao
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Weili Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yawen Zhang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinchi Jiang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qianbin Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanyan Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing 100029, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, Beijing 100029, China
- Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yong Wang
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
- Beijing Key Laboratory of TCM Syndrome and Formula, Beijing 100029, China
- Key Laboratory of Beijing University of Chinese Medicine, Ministry of Education, Beijing 100029, China
| |
Collapse
|
13
|
Abrahams C, Woudberg NJ, Lecour S. Anthracycline-induced cardiotoxicity: targeting high-density lipoproteins to limit the damage? Lipids Health Dis 2022; 21:85. [PMID: 36050733 PMCID: PMC9434835 DOI: 10.1186/s12944-022-01694-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/02/2022] [Indexed: 12/30/2022] Open
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic frequently used against a wide range of cancers, including breast cancer. Although the drug is effective as a treatment against cancer, many patients develop heart failure (HF) months to years following their last treatment with DOX. The challenge in preventing DOX-induced cardiotoxicity is that symptoms present after damage has already occurred in the myocardium. Therefore, early biomarkers to assess DOX-induced cardiotoxicity are urgently needed. A better understanding of the mechanisms involved in the toxicity is important as this may facilitate the development of novel early biomarkers or therapeutic approaches. In this review, we discuss the role of high-density lipoprotein (HDL) particles and its components as possible key players in the early development of DOX-induced cardiotoxicity. HDL particles exist in different subclasses which vary in composition and biological functionality. Multiple cardiovascular risk factors are associated with a change in HDL subclasses, resulting in modifications of their composition and physiological functions. There is growing evidence in the literature suggesting that cancer affects HDL subclasses and that healthy HDL particles enriched with sphingosine-1-phosphate (S1P) and apolipoprotein A1 (ApoA1) protect against DOX-induced cardiotoxicity. Here, we therefore discuss associations and relationships between HDL, DOX and cancer and discuss whether assessing HDL subclass/composition/function may be considered as a possible early biomarker to detect DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Carmelita Abrahams
- Cardioprotection Group, Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, 7935, South Africa
| | - Nicholas J Woudberg
- Cardioprotection Group, Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, 7935, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, 7935, South Africa.
| |
Collapse
|
14
|
Mohan UP, Tirupathi Pichiah PB, Kunjiappan S, Arunachalam S. A Hypothesis Concerning the role of PPAR family on Cardiac Energetics in Adriamycin-Induced Cardiomyopathy. J Appl Toxicol 2022; 42:1910-1920. [PMID: 35944906 DOI: 10.1002/jat.4374] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/06/2022] [Accepted: 08/06/2022] [Indexed: 11/10/2022]
Abstract
Adriamycin is an effective anti-neoplastic drug against a variety of cancer types. However, the drug causes adverse side-effects in a number of organ systems. Cardiomyopathy is one of the life-threatening side-effects of Adriamycin. In the current work, we have derived the possible involvement of PPAR family members in the development of Adriamycin-induced cardiomyopathy. Dysregulation of PPAR family by Adriamycin causes impairment in the transport and β-oxidation of fatty acids, the key substrate for ATP synthesis in heart. Evidences suggest that dysregulation of PPAR family results in alters the recruitment of glucose transporters. Furthermore, Hemeoxygenase-1 is a crucial enzyme regulating the iron homeostasis in the heart whose expression is regulated by PPAR family. Inverse relationship exists between the expression levels of PPARγ and hemeoxygenase-1. Adriamycin upregulates the expression of hemeoxygenase-1 which in turn disrupts the iron homeostasis in cardiomyocytes. Our molecular docking results show that Adriamycin has high affinity for iron binding sites of hemeoxygenase-1, thereby hindering formation of iron-sulfur complex. Lack of iron-sulfur complex impairs the electron transport chain. In addition, succinate dehydrogenase subunit A is downregulated by Adriamycin. The lack of this subunit uncouples Krebs cycle from ETC. Further lack of this subunit causes increases the concentration of succinate which further alters the mitochondrial membrane potential. Overall, in the present work we hypothesize that alteration in the expression of PPAR family members is one of the major causes of metabolic chaos and oxidative stress caused by Adriamycin during the development of cardiomyopathy.
Collapse
Affiliation(s)
- Uma Priya Mohan
- Centre for Cardiovascular and Adverse Drug Reactions, Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, India
| | | | - Selvaraj Kunjiappan
- Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, India
| | - Sankarganesh Arunachalam
- Centre for Cardiovascular and Adverse Drug Reactions, Department of Biotechnology, School of Bio and Chemical Engineering, Kalasalingam Academy of Research and Education, Krishnankoil, Tamilnadu, India
| |
Collapse
|
15
|
Dourado MLC, Dompieri LT, Leitão GM, Mourato FA, Santos RGG, Almeida PJ, Markman B, Melo MDT, Brandão SCS. Aumento de Captação Cardíaca de 18F-FDG Induzida por Quimioterapia em Pacientes com Linfoma: Um Marcador Precoce de Cardiotoxicidade? Arq Bras Cardiol 2022; 118:1049-1058. [PMID: 35703659 PMCID: PMC9345149 DOI: 10.36660/abc.20210463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/01/2021] [Indexed: 11/18/2022] Open
Abstract
Fundamento Ainda não está estabelecido se a captação de fluorodesoxiglicose no miocárdio ocorre exclusivamente por características fisiológicas ou se representa um desarranjo metabólico causado pela quimioterapia. Objetivo Investigar os efeitos da quimioterapia no coração dos pacientes com linfoma por tomografia por emissão de pósitrons associada a tomografia computadorizada (PET/CT) com 2-[18F]-fluoro-2-desoxi-D-glicose (18F-FDG PET/CT) antes, durante e/ou após a quimioterapia. Métodos Setenta pacientes com linfoma submetidos a 18F-FDG PET/CT foram retrospectivamente analisados. O nível de significância foi de 5%. A captação de 18F-FDG foi avaliada por três medidas: captação máxima no ventrículo esquerdo ( standardized uptake value , SUV max), razão SUV cardíaco / aorta e SUV cardíaco / SUV no fígado. Também foram comparados peso corporal, glicemia de jejum, tempo pós-injeção e dose administrada de 18F-FDG entre os exames. Resultados A idade média foi de 50,4 ± 20,1 anos e 50% dos pacientes eram mulheres. A análise foi realizada em dois grupos – PET/CT basal vs. intermediário e PET/CT basal vs pós-terapia. Não houve diferença significativa entre as variáveis clínicas e do protocolo dos exames entre os diferentes momentos avaliados. Nós observamos um aumento na SUV máxima no ventrículo esquerdo de 3,5±1,9 (basal) para 5,6±4,0 (intermediário), p=0,01, e de 4,0±2,2 (basal) para 6,1±4,2 (pós-terapia), p<0,001. Uma porcentagem de aumento ≥30% na SUV máxima no ventrículo esquerdo ocorreu em mais da metade da amostra. O aumento da SUV cardíaca foi acompanhado por um aumento na razão SUV máxima no ventrículo esquerdo / SUV máxima na aorta e SUV média no ventrículo esquerdo /SUV média no fígado. Conclusão O estudo mostrou um aumento evidente na captação cardíaca de 18F-FDG em pacientes com linfoma, durante e após quimioterapia. A literatura corrobora com esses achados e sugere que a 18F-FDG PET/CT pode ser um exame de imagem sensível e confiável para detectar sinais metabólicos precoces de cardiotoxicidade.
Collapse
|
16
|
Wang H, Yu X, Xun Z, Wu Y. Aqueous Extract of Andrographis paniculata Ameliorates Cardiotoxicity Induced by Doxorubicin in vivo. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.466.474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
17
|
Nauclea orientalis (L.) Bark Extract Protects Rat Cardiomyocytes from Doxorubicin-Induced Oxidative Stress, Inflammation, Apoptosis, and DNA Fragmentation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1714841. [PMID: 35198093 PMCID: PMC8860544 DOI: 10.1155/2022/1714841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/22/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022]
Abstract
The therapeutic efficacy of anthracycline antibiotic, doxorubicin (Dox), is hampered due to the dose-dependent cardiotoxicity. The objective of the study was to explore the counteraction of aqueous bark extract of Nauclea orientalis in Dox-induced cardiotoxicity in Wistar rats. The acute and subchronic toxicity study performed with 2.0 g/kg of the plant extract revealed biochemical and haematological parameters to be within the physiological range, and no histological alterations were observed in any organs isolated. Screening of plant extract for the protection of the myocardium from Dox-induced oxidative stress, inflammation, and apoptosis was performed on five groups of rats: control, plant extract control, Dox control (distilled water (D.H2O) 2 weeks + on the 11th day single injection of Dox, 18 mg/kg), plant + Dox (2.0 g/kg plant extract 2 weeks + on the 11th day Dox, 18 mg/kg), and positive control, dexrazoxane. A significant increase in cardiac biomarkers and lipid peroxidation (p < 0.001) and a significant decrease in antioxidant parameters (p < 0.001) were observed in the Dox control group. All these parameters were reversed significantly (p < 0.05) in the plant-pretreated group. The histopathological assessment of myocardial damage provided supportive evidence for the biochemical results obtained. Inflammatory markers, myeloperoxidase, expression of TNFα and caspase-3, and DNA fragmentation (TUNEL positive nuclei) were significantly elevated (p < 0.05), and expression of Bcl-2 was significantly decreased (p < 0.05) in the Dox control; however, all these parameters were significantly reversed in the plant extract-treated group. In conclusion, the aqueous bark extract of Nauclea orientalis (2.0 g/kg) has the ability to attenuate the Dox-induced oxidative stress, inflammation, apoptosis, and DNA fragmentation in Wistar rats.
Collapse
|
18
|
Selyutina OY, Kononova PA, Koshman VE, Fedenok LG, Polyakov NE. The Interplay of Ascorbic Acid with Quinones-Chelators—Influence on Lipid Peroxidation: Insight into Anticancer Activity. Antioxidants (Basel) 2022; 11:antiox11020376. [PMID: 35204258 PMCID: PMC8869476 DOI: 10.3390/antiox11020376] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/24/2022] [Accepted: 02/11/2022] [Indexed: 12/11/2022] Open
Abstract
Ascorbic acid is a multifaceted compound that can perform both antioxidant and pro-oxidant activities in the redox reactions induced by transition metal ions, so its role in nature and especially in the human body is still the subject of debate. In the present study, we have examined the influence of ascorbic acid on lipid peroxidation in a model system that mimics the cell membrane, namely micelles of linoleic acid (LA), induced by chelate complexes of iron and copper ions with quinone-chelator 2-phenyl-4-(butylamino)-naphtholquinoline-7,12-dione (Q1). This quinone effectively generates reactive oxygen species and semiquinone radicals inside cancer cells via a cycling redox reaction. Here it was demonstrated that in the absence of quinone-chelator ascorbic acid significantly accelerates the lipid peroxidation induced by both Fe(II) and Cu(II) ions. It has been shown also that Q1 chelate complexes with Fe(II) and Cu(II) ions are redox active in the LA micelles oxidation. No effect of ascorbate was detected on the reactivity of chelate complex with Fe(II) ions. On the other hand, ascorbate performs pro-oxidant activity in Q1-Cu(II) complex induced reaction. We can conclude that ascorbate-driven redox cycling of Q1 may promote its anti-tumor activity.
Collapse
|
19
|
Huang J, Wu R, Chen L, Yang Z, Yan D, Li M. Understanding Anthracycline Cardiotoxicity From Mitochondrial Aspect. Front Pharmacol 2022; 13:811406. [PMID: 35211017 PMCID: PMC8861498 DOI: 10.3389/fphar.2022.811406] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/24/2022] [Indexed: 01/18/2023] Open
Abstract
Anthracyclines, such as doxorubicin, represent one group of chemotherapy drugs with the most cardiotoxicity. Despite that anthracyclines are capable of treating assorted solid tumors and hematological malignancies, the side effect of inducing cardiac dysfunction has hampered their clinical use. Currently, the mechanism underlying anthracycline cardiotoxicity remains obscure. Increasing evidence points to mitochondria, the energy factory of cardiomyocytes, as a major target of anthracyclines. In this review, we will summarize recent findings about mitochondrial mechanism during anthracycline cardiotoxicity. In particular, we will focus on the following aspects: 1) the traditional view about anthracycline-induced reactive oxygen species (ROS), which is produced by mitochondria, but in turn causes mitochondrial injury. 2) Mitochondrial iron-overload and ferroptosis during anthracycline cardiotoxicity. 3) Autophagy, mitophagy and mitochondrial dynamics during anthracycline cardiotoxicity. 4) Anthracycline-induced disruption of cardiac metabolism.
Collapse
Affiliation(s)
- Junqi Huang
- Key Laboratory for Regenerative Medicine, Ministry of Education, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Rundong Wu
- Department of Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Linyi Chen
- Department of Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Ziqiang Yang
- Department of Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Daoguang Yan
- Department of Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Mingchuan Li
- Department of Biology, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
20
|
Dependence of glucose transport on autophagy and GAPDH activity. Brain Res 2022; 1776:147747. [PMID: 34864044 PMCID: PMC8819679 DOI: 10.1016/j.brainres.2021.147747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/20/2021] [Accepted: 11/29/2021] [Indexed: 02/03/2023]
Abstract
Glucose uptake in the brain is critically important to brain health. Using two widely used cell line model systems, we have found that siramesine, a lysosomotropic agent and ligand for the sigma-2 receptor, inhibits glucose uptake and decreases pools of the GLUT1 glucose transporter at the plasma membrane. Siramesine induces autophagy but also disrupts degradation of autophagy substrates, providing a potential mechanism for its action on glucose uptake. In other cell systems, many of the effects of siramesine can be suppressed by α -tocopherol, a type of vitamin E and potent antioxidant, and α-tocopherol also suppressed the effect of siramesine on glucose uptake, suggesting a role for reactive oxygen species and membrane maintenance. We have also identified a novel mechanism for siramesine in which it inhibited plasma membrane levels of GAPDH, a key protein in glycolysis which localizes to the plasma membrane in some cell types. Indeed, GAPDH inhibitors decreased glucose uptake, like siramesine, likely through an overlapping pathway with siramesine. GAPDH inhibitors induced autophagy but inhibited degradation of autophagy targets. Thus, we have identified novel mechanisms required for glucose uptake which may have important implications in disease.
Collapse
|
21
|
Mechanistic Insights of Chelator Complexes with Essential Transition Metals: Antioxidant/Pro-Oxidant Activity and Applications in Medicine. Int J Mol Sci 2022; 23:ijms23031247. [PMID: 35163169 PMCID: PMC8835618 DOI: 10.3390/ijms23031247] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
The antioxidant/pro-oxidant activity of drugs and dietary molecules and their role in the maintenance of redox homeostasis, as well as the implications in health and different diseases, have not yet been fully evaluated. In particular, the redox activity and other interactions of drugs with essential redox metal ions, such as iron and copper, need further investigation. These metal ions are ubiquitous in human nutrition but also widely found in dietary supplements and appear to exert major effects on redox homeostasis in health, but also on many diseases of free radical pathology. In this context, the redox mechanistic insights of mainly three prototype groups of drugs, namely alpha-ketohydroxypyridines (alpha-hydroxypyridones), e.g., deferiprone, anthraquinones, e.g., doxorubicin and thiosemicarbazones, e.g., triapine and their metal complexes were examined; details of the mechanisms of their redox activity were reviewed, with emphasis on the biological implications and potential clinical applications, including anticancer activity. Furthermore, the redox properties of these three classes of chelators were compared to those of the iron chelating drugs and also to vitamin C, with an emphasis on their potential clinical interactions and future clinical application prospects in cancer, neurodegenerative and other diseases.
Collapse
|
22
|
Thompson EW, Demissei BG, Smith AM, Brahmbhatt P, Wang J, Clark A, DeMichele A, Narayan V, Shah P, Sun L, Lefebvre B, Fradley MG, Carver JR, Tang WW, Ky B. Paraoxonase-1 Activity in Breast Cancer Patients Treated With Doxorubicin With or Without Trastuzumab. JACC Basic Transl Sci 2022; 7:1-10. [PMID: 35128203 PMCID: PMC8807731 DOI: 10.1016/j.jacbts.2021.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 11/15/2022]
Abstract
PON-1 is an HDL-associated cardioprotective enzyme that prevents oxidized-LDL formation and has not previously been studied in cardio-oncology. To determine the associations between PON-1 and the development of CTRCD, the Pon and Aryl serum enzymatic activity levels of PON-1 were quantified in a cohort of 225 patients with breast cancer receiving doxorubicin with or without trastuzumab. After doxorubicin completion, the activity levels of both Pon and Aryl were significantly decreased. Early increases in the Pon enzymatic activity of PON-1 were associated with increased risk of CTRCD. With further study, PON-1 activity may provide insight into mechanistic risk prediction of CTRCD with doxorubicin chemotherapy.
The objective of this study was to determine associations of paraoxonase-1 (PON-1) with development of cancer therapy–related cardiac dysfunction (CTRCD). PON-1 is a cardioprotective enzyme associated with high-density lipoprotein that prevents oxidized low-density lipoprotein formation. Given the role of oxidative stress in doxorubicin-induced cardiotoxicity, PON-1 activity may have relevance for the prediction of CTRCD. In 225 patients with breast cancer receiving doxorubicin with or without trastuzumab, we quantified PON-1 activity through its paraoxonase (Pon) and arylesterase (Aryl) enzymatic activity at baseline, during, and after doxorubicin completion. Echocardiograms were performed at baseline, during therapy, and annually. CTRCD was defined as a decrease in left ventricular ejection fraction by ≥10% from baseline to <50%. Associations between baseline biomarkers and clinical variables were determined using multivariable linear regression. Associations between changes in biomarker activity and time to CTRCD were evaluated using Cox regression. Pon was directly associated with Black race and inversely associated with Stage 2 cancer. Aryl was inversely associated with body mass index. After doxorubicin completion, activity levels of Pon and Aryl were significantly decreased (median ratio compared with baseline for Pon: 0.95 [Q1-Q3: 0.81-1.07, P < 0.001]; for Aryl: 0.97 [Q1-Q3: 0.85-1.08, P = 0.010]). A total of 184 patients had an available quantitated echocardiogram at baseline and at least 1 follow-up visit. Increases from baseline in Pon at doxorubicin completion were independently associated with increased CTRCD risk (per 10% increase: hazard ratio [HR]: 1.21; 95% confidence interval [CI]: 1.05-1.39; P = 0.007). Associations between increases in Aryl and CTRCD tended in the same direction but were of borderline statistical significance (HR: 1.17; 95% CI: 0.99-1.38; P = 0.071). In patients with breast cancer treated with doxorubicin with or without trastuzumab, increases in the Pon enzymatic activity level of PON-1 were associated with increased CTRCD risk. PON-1 activity may be relevant to mechanistic risk prediction of cardiotoxicity with anthracyclines.
Collapse
Key Words
- Aryl, arylesterase
- BMI, body mass index
- CTRCD, cancer therapy–related cardiac dysfunction
- CVD, cardiovascular disease
- HDL, high-density lipoprotein
- HER2, human epidermal growth factor receptor 2
- LDL, low-density lipoprotein
- LVEF, left ventricular ejection fraction
- PON-1, paraoxonase-1
- Pon, paraoxonase
- cardiac dysfunction
- cardiotoxicity
- doxorubicin
- heart failure
- paraoxonase-1
Collapse
Affiliation(s)
- Elizabeth W. Thompson
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Biniyam G. Demissei
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amanda M. Smith
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Priya Brahmbhatt
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jessica Wang
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amy Clark
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Angela DeMichele
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Vivek Narayan
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Payal Shah
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lova Sun
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benedicte Lefebvre
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michael G. Fradley
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph R. Carver
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - W.H. Wilson Tang
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Address for correspondence: Dr Bonnie Ky, Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Office 11-105 Smilow Center for Translational Research, 3400 Civic Center Boulevard, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
23
|
Quagliariello V, De Laurentiis M, Rea D, Barbieri A, Monti MG, Carbone A, Paccone A, Altucci L, Conte M, Canale ML, Botti G, Maurea N. The SGLT-2 inhibitor empagliflozin improves myocardial strain, reduces cardiac fibrosis and pro-inflammatory cytokines in non-diabetic mice treated with doxorubicin. Cardiovasc Diabetol 2021; 20:150. [PMID: 34301253 PMCID: PMC8305868 DOI: 10.1186/s12933-021-01346-y] [Citation(s) in RCA: 235] [Impact Index Per Article: 58.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/16/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Empagliflozin (EMPA), a selective inhibitor of the sodium glucose co-transporter 2, reduced the risk of hospitalization for heart failure and cardiovascular death in type 2 diabetic patients in the EMPA-REG OUTCOME trial. Recent trials evidenced several cardio-renal benefits of EMPA in non-diabetic patients through the involvement of biochemical pathways that are still to be deeply analysed. We aimed to evaluate the effects of EMPA on myocardial strain of non-diabetic mice treated with doxorubicin (DOXO) through the analysis of NLRP3 inflammasome and MyD88-related pathways resulting in anti-apoptotic and anti-fibrotic effects. METHODS Preliminary cellular studies were performed on mouse cardiomyocytes (HL-1 cell line) exposed to doxorubicin alone or combined to EMPA. The following analysis were performed: determination of cell viability (through a modified MTT assay), study of intracellular ROS production, lipid peroxidation (quantifying intracellular malondialdehyde and 4-hydroxynonenal), intracellular Ca2+ homeostasis. Moreover, pro-inflammatory studies were also performed: expression of NLRP3 inflammasome, MyD88 myddosome and p65/NF-κB associated to secretion of cytokines involved in cardiotoxicity (Interleukins 1β, 8, 6). C57Bl/6 mice were untreated (Sham, n = 6) or treated for 10 days with doxorubicin (DOXO, n = 6), EMPA (EMPA, n = 6) or doxorubicin combined to EMPA (DOXO-EMPA, n = 6). DOXO was injected intraperitoneally. Ferroptosis and xanthine oxidase were studied before and after treatments. Cardiac function studies, including EF, FS and radial/longitudinal strain were analysed through transthoracic echocardiography (Vevo 2100). Cardiac fibrosis and apoptosis were histologically studied through Picrosirius red and TUNEL assay, respectively and quantified through pro-collagen-1α1, MMP-9 and Caspase-3 expression. Tissue NLRP3, MyD88 and cytokines were also quantified before and after treatments through ELISA methods. RESULTS Cardiomyocytes exposed to doxorubicin increased the intracellular Ca2+ content and expression of several pro-inflammatory markers associated to cell death; co-incubation with EMPA reduced significantly the magnitude of the effects. In preclinical study, EMPA increased EF and FS compared to DOXO groups (p < 0.05), prevented the reduction of radial and longitudinal strain after 10 days of treatment with doxorubicin (RS) 30.3% in EMPA-DOXO vs 15.7% in DOXO mice; LS - 17% in EMPA-DOXO vs - 11.7% in DOXO mice (p < 0.001 for both). Significant reductions in ferroptosis, xanthine oxidase expression, cardiac fibrosis and apoptosis in EMPA associated to DOXO were also seen. A reduced expression of pro-inflammatory cytokines, NLRP3, MyD88 and NF-kB in heart, liver and kidneys was also seen in DOXO-EMPA group compared to DOXO (p < 0.001). CONCLUSION EMPA reduced ferroptosis, fibrosis, apoptosis and inflammation in doxorubicin-treated mice through the involvement of NLRP3 and MyD88-related pathways, resulting in significant improvements in cardiac functions. These findings provides the proof of concept for translational studies designed to reduce adverse cardiovascular outcomes in non-diabetic cancer patients treated with doxorubicin.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy.
| | | | - Domenica Rea
- SSD Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Antonio Barbieri
- SSD Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Maria Gaia Monti
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Andreina Carbone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Andrea Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Mariarosaria Conte
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Maria Laura Canale
- Cardiology Division, Azienda USL Toscana Nord-Ovest, Versilia Hospital, Lido Di Camaiore, Italy
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
24
|
Córdoba-Adaya JC, Oros-Pantoja R, Torres-García E, Morales-Ávila E, Aranda-Lara L, Santillán-Benítez JG, Hernández-Herrera NO, Otero G, Isaac-Olivé K. Evaluation of doxorubicin-induced early multi-organ toxicity in male CD1 mice by biodistribution of 18F-FDG and 67Ga-citrate. Pilot study. Toxicol Mech Methods 2021; 31:546-558. [PMID: 34057017 DOI: 10.1080/15376516.2021.1937420] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The search for methods that identify early toxicity, induced by chemotherapy, is urgent. Changes in the biodistribution of radiopharmaceuticals could give information on early toxicity. Ten-week-old CD1 male mice were divided into four groups. Two groups were administered a weekly dose of 5 mg/kg of doxorubicin hydrochloride (DOX) for 5 weeks and the control groups were administered saline solution. One week after the end of treatment, the biodistribution of 18F-FDG and 67Ga-citrate were carried out, as was the quantification of plasma enzymes CK, CK-MB, LDH and AST. All enzymes were higher in the treated animals, but only significant (p < 0.05) in the case of CK-MB. 18F-FDG uptake increased in all organs of treated animals except retroperitoneal fat, being significant in spleen, brain, heart, liver, lung, kidney, and inguinal fat. 67Ga-citrate had a more complex pattern. The uptake in the DOX group was higher in spleen, lung, kidney, testes, and gonadal fat, it did not change in brain, heart, and liver, and it was lower in the rest of the organs. It only showed significant differences in lung and pancreas. A thorough discussion of the possible causes that produced the change in biodistributions of both radiopharmaceuticals is included. The pilot study showed that both radiopharmaceuticals could identify early multi-organ toxicity induced by DOX. Although 18F-FDG seems to be better, 67Ga-citrato should not be ruled out a priori. The detection of early toxicity would serve to adopt treatments that prevent its progression, thus improving patient's quality of life.
Collapse
Affiliation(s)
- Julio César Córdoba-Adaya
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Rigoberto Oros-Pantoja
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Eugenio Torres-García
- Laboratorio de Dosimetría y Simulación Monte Carlo, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Enrique Morales-Ávila
- Laboratorio de Farmacia y Toxicología, Facultad de Química, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Liliana Aranda-Lara
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Jonnathan G Santillán-Benítez
- Laboratorio de Farmacia y Toxicología, Facultad de Química, Universidad Autónoma del Estado de México, Toluca, Mexico
| | | | - Gloria Otero
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Keila Isaac-Olivé
- Laboratorio de Investigación en Teranóstica, Facultad de Medicina, Universidad Autónoma del Estado de México, Toluca, Mexico
| |
Collapse
|
25
|
Minamimoto R. Series of myocardial FDG uptake requiring considerations of myocardial abnormalities in FDG-PET/CT. Jpn J Radiol 2021; 39:540-557. [PMID: 33517516 PMCID: PMC8175248 DOI: 10.1007/s11604-021-01097-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/16/2021] [Indexed: 12/18/2022]
Abstract
Distinct from cardiac PET performed with preparation to control physiological FDG uptake in the myocardium, standard FDG-PET/CT performed with 4-6 h of fasting will show variation in myocardial FDG uptake. For this reason, important signs of myocardial and pericardial abnormality revealed by myocardial FDG uptake tend to be overlooked. However, recognition of possible underlying disease will support further patient management to avoid complications due to the disease. This review demonstrates the mechanism of FDG uptake in the myocardium, discusses the factors affecting uptake, and provides notable image findings that may suggest underlying disease.
Collapse
Affiliation(s)
- Ryogo Minamimoto
- Division of Nuclear Medicine, Department of Radiology, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjyuku-ku, Tokyo, 162-8655, Japan.
| |
Collapse
|
26
|
Aziz TA. Cardioprotective Effect of Quercetin and Sitagliptin in Doxorubicin-Induced Cardiac Toxicity in Rats. Cancer Manag Res 2021; 13:2349-2357. [PMID: 33737832 PMCID: PMC7965691 DOI: 10.2147/cmar.s300495] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Objective A previous study revealed a pronounced protective effect of combining quercetin (QC) with sitagliptin (STN) in testicular tissue. Accordingly, this study was designed to evaluate the cardioprotective effects of QC and STN each alone or in combination in doxorubicin (DOX)-induced cardiotoxicity in the rats. Methodology Thirty male adult Wistar rats were divided into five groups: the first group (control) treated with sodium chloride, the second group treated with DOX (3 mg/kg I.P. injection), the third group treated with DOX with a combination of QC (80 mg/kg), and STN (10 mg/kg), the fourth group treated with DOX and QC and the fifth group treated with DOX and STN. Blood was collected on day 22 and used for assessment of serum troponin, lactate dehydrogenase (LDH), creatine phosphokinase (CPK), total lipid profile, C-reactive protein (CRP), and total antioxidant capacity (TAOC). Atherogenic indices were also calculated. Cardiac tissue was sent for histopathological analysis. Results DOX produced a significant increase in the level of troponin, LDH, CKP, CRP, total cholesterol (TC), low-density lipoprotein (LDL), triglycerides (TG), and atherogenic index of plasma; and significantly decreased TAOC. The combination of quercetin and sitagliptin was more effective than each treatment alone in restoring the level of troponin, LDH, CKP, CRP, Cholesterol, LDL, TG, atherogenic index of plasma and significantly increased TAOC compared to DOX treated group. The histopathological finding also supports the biochemical results. Conclusion The study revealed the cardioprotective effects of the combination of QC and STN which could be attributed to the additive effects of this combination through antioxidant, anti-inflammatory, lipid lowering and anti-atherogenic activities; suggesting it as a good therapeutic candidate to be tested in the clinical setting.
Collapse
Affiliation(s)
- Tavga Ahmed Aziz
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Sulaimani, Sulaimani City, Iraq
| |
Collapse
|
27
|
Russo M, Della Sala A, Tocchetti CG, Porporato PE, Ghigo A. Metabolic Aspects of Anthracycline Cardiotoxicity. Curr Treat Options Oncol 2021; 22:18. [PMID: 33547494 PMCID: PMC7864817 DOI: 10.1007/s11864-020-00812-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 12/13/2022]
Abstract
OPINION STATEMENT Heart failure (HF) is increasingly recognized as the major complication of chemotherapy regimens. Despite the development of modern targeted therapies such as monoclonal antibodies, doxorubicin (DOXO), one of the most cardiotoxic anticancer agents, still remains the treatment of choice for several solid and hematological tumors. The insurgence of cardiotoxicity represents the major limitation to the clinical use of this potent anticancer drug. At the molecular level, cardiac side effects of DOXO have been associated to mitochondrial dysfunction, DNA damage, impairment of iron metabolism, apoptosis, and autophagy dysregulation. On these bases, the antioxidant and iron chelator molecule, dexrazoxane, currently represents the unique FDA-approved cardioprotectant for patients treated with anthracyclines.A less explored area of research concerns the impact of DOXO on cardiac metabolism. Recent metabolomic studies highlight the possibility that cardiac metabolic alterations may critically contribute to the development of DOXO cardiotoxicity. Among these, the impairment of oxidative phosphorylation and the persistent activation of glycolysis, which are commonly observed in response to DOXO treatment, may undermine the ability of cardiomyocytes to meet the energy demand, eventually leading to energetic failure. Moreover, increasing evidence links DOXO cardiotoxicity to imbalanced insulin signaling and to cardiac insulin resistance. Although anti-diabetic drugs, such as empagliflozin and metformin, have shown interesting cardioprotective effects in vitro and in vivo in different models of heart failure, their mechanism of action is unclear, and their use for the treatment of DOXO cardiotoxicity is still unexplored.This review article aims at summarizing current evidence of the metabolic derangements induced by DOXO and at providing speculations on how key players of cardiac metabolism could be pharmacologically targeted to prevent or cure DOXO cardiomyopathy.
Collapse
Affiliation(s)
- Michele Russo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Via Nizza 52, 10126, Torino, Italy
| | - Angela Della Sala
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Via Nizza 52, 10126, Torino, Italy
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Paolo Ettore Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Via Nizza 52, 10126, Torino, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
28
|
Xie L, Wang G, Sang W, Li J, Zhang Z, Li W, Yan J, Zhao Q, Dai Y. Phenolic immunogenic cell death nanoinducer for sensitizing tumor to PD-1 checkpoint blockade immunotherapy. Biomaterials 2020; 269:120638. [PMID: 33421711 DOI: 10.1016/j.biomaterials.2020.120638] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 12/21/2020] [Accepted: 12/27/2020] [Indexed: 12/16/2022]
Abstract
A critical challenge remains in PD-1 checkpoint blockade immunotherapy is few tumor specific T cells infiltration in hypoxic tumor microenvironment (TME). Improving immunogenic cell death (ICD) associated immunogenicity can make tumor sensitive to PD-1 checkpoint blockade immunotherapy. Herein, a phenolic ICD inducer was engineered by self-assembly of the superior ICD inducer (doxorubicin, DOX), phenolic manganese dioxide nanoreactor, ferric iron and PEG-polyphenols (MDP NPs) via metal phenolic coordination. These oxygen self-supporting MDP NPs strengthen DOX based ROS-dependent cell death and their metal mediated chemodynamic effect accelerate ICD induction. Together with concomitant ICD triggered by DOX, MDP NPs successively lead to tumor-associated antigen boosting, DCs maturation and ultimately enhance tumor-specific T cells infiltration. Furthermore, MDP NPs efficiently modulated hypoxic TME for effective macrophages recruitment. This promising ICD-augment strategy efficiently improve tumor response to PD-1 checkpoint blockade immunotherapy, resulting in a significant antitumor immune response in primary tumor and a strong abscopal effect to distant tumor. Our simple and versatile phenolic inducer expands the application of chemodrugs based ICD enhancing PD-1 checkpoint blockade immunotherapy.
Collapse
Affiliation(s)
- Lisi Xie
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Guohao Wang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Wei Sang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Jie Li
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Zhan Zhang
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Wenxi Li
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Jie Yan
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Qi Zhao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Yunlu Dai
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China; Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China.
| |
Collapse
|
29
|
Narzt MS, Pils V, Kremslehner C, Nagelreiter IM, Schosserer M, Bessonova E, Bayer A, Reifschneider R, Terlecki-Zaniewicz L, Waidhofer-Söllner P, Mildner M, Tschachler E, Cavinato M, Wedel S, Jansen-Dürr P, Nanic L, Rubelj I, El-Ghalbzouri A, Zoratto S, Marchetti-Deschmann M, Grillari J, Gruber F, Lämmermann I. Epilipidomics of Senescent Dermal Fibroblasts Identify Lysophosphatidylcholines as Pleiotropic Senescence-Associated Secretory Phenotype (SASP) Factors. J Invest Dermatol 2020; 141:993-1006.e15. [PMID: 33333126 DOI: 10.1016/j.jid.2020.11.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
During aging, skin accumulates senescent cells. The transient presence of senescent cells, followed by their clearance by the immune system, is important in tissue repair and homeostasis. The persistence of senescent cells that evade clearance contributes to the age-related deterioration of the skin. The senescence-associated secretory phenotype of these cells contains immunomodulatory molecules that facilitate clearance but also promote chronic damage. Here, we investigated the epilipidome-the oxidative modifications of phospholipids-of senescent dermal fibroblasts, because these molecules are among the bioactive lipids that were recently identified as senescence-associated secretory phenotype factors. Using replicative- and stress- induced senescence protocols, we identified lysophosphatidylcholines as universally elevated in senescent fibroblasts, whereas other oxidized lipids displayed a pattern that was characteristic for the used senescence protocol. When we tested the lysophosphatidylcholines for senescence-associated secretory phenotype activity, we found that they elicit chemokine release in nonsenescent fibroblasts but also interfere with toll-like receptor 2 and 6/CD36 signaling and phagocytic capacity in macrophages. Using matrix-assisted laser desorption/ionization Fourier transform ion cyclotron resonance mass spectrometry imaging, we localized two lysophosphatidylcholine species in aged skin. This suggests that lysophospholipids may facilitate immune evasion and low-grade chronic inflammation in skin aging.
Collapse
Affiliation(s)
- Marie-Sophie Narzt
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz and Vienna, Austria
| | - Vera Pils
- Department of Dermatology, Medical University of Vienna, Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Christopher Kremslehner
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria
| | - Ionela-Mariana Nagelreiter
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria; Center for Brain Research, Department of Molecular Neurosciences, Medical University of Vienna, Vienna, Austria
| | - Markus Schosserer
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Emilia Bessonova
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Alina Bayer
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Raffaela Reifschneider
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Lucia Terlecki-Zaniewicz
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Petra Waidhofer-Söllner
- Institute of Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Erwin Tschachler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Maria Cavinato
- Institute for Biomedical Aging Research, University of Innsbruck, Austria; Center for Molecular Biosciences Innsbruck, Innsbruck, Austria
| | - Sophia Wedel
- Institute for Biomedical Aging Research, University of Innsbruck, Austria; Center for Molecular Biosciences Innsbruck, Innsbruck, Austria
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, University of Innsbruck, Austria; Center for Molecular Biosciences Innsbruck, Innsbruck, Austria
| | - Lucia Nanic
- Ruder Boskovic Institute, Division of Molecular Biology, Laboratory for Molecular and Cellular Biology, Zagreb, Croatia
| | - Ivica Rubelj
- Ruder Boskovic Institute, Division of Molecular Biology, Laboratory for Molecular and Cellular Biology, Zagreb, Croatia
| | | | - Samuele Zoratto
- Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria; Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Martina Marchetti-Deschmann
- Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Institute of Chemical Technologies and Analytics, TU Wien, Vienna, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, Linz and Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Florian Gruber
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Dermatology, Medical University of Vienna, Vienna, Austria; Christian Doppler Laboratory for Skin Multimodal Imaging of Aging and Senescence, Vienna, Austria.
| | - Ingo Lämmermann
- Christian Doppler Laboratory on Biotechnology of Skin Aging, Vienna, Austria; Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| |
Collapse
|
30
|
Bauckneht M, Pastorino F, Castellani P, Cossu V, Orengo AM, Piccioli P, Emionite L, Capitanio S, Yosifov N, Bruno S, Lazzarini E, Ponzoni M, Ameri P, Rubartelli A, Ravera S, Morbelli S, Sambuceti G, Marini C. Increased myocardial 18F-FDG uptake as a marker of Doxorubicin-induced oxidative stress. J Nucl Cardiol 2020; 27:2183-2194. [PMID: 30737636 DOI: 10.1007/s12350-019-01618-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/07/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Oxidative stress and its interference on myocardial metabolism play a major role in Doxorubicin (DXR) cardiotoxic cascade. METHODS Mice models of neuroblastoma (NB) were treated with 5 mg DXR/kg, either free (Free-DXR) or encapsulated in untargeted (SL[DXR]) or in NB-targeting Stealth Liposomes (pep-SL[DXR] and TP-pep-SL[DXR]). Control mice received saline. FDG-PET was performed at baseline (PET1) and 7 days after therapy (PET2). At PET2 Troponin-I and NT-proBNP were assessed. Explanted hearts underwent biochemical, histological, and immunohistochemical analyses. Finally, FDG uptake and glucose consumption were simultaneously measured in cultured H9c2 in the presence/absence of Free-DXR (1 μM). RESULTS Free-DXR significantly enhanced the myocardial oxidative stress. Myocardial-SUV remained relatively stable in controls and mice treated with liposomal formulations, while it significantly increased at PET2 with respect to baseline in Free-DXR. At this timepoint, myocardial-SUV was directly correlated with both myocardial redox stress and hexose-6-phosphate-dehydrogenase (H6PD) enzymatic activity, which selectively sustain cellular anti-oxidant mechanisms. Intriguingly, in vitro, Free-DXR selectively increased FDG extraction fraction without altering the corresponding value for glucose. CONCLUSION The direct correlation between cardiac FDG uptake and oxidative stress indexes supports the potential role of FDG-PET as an early biomarker of DXR oxidative damage.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
- Nuclear Medicine, Department of Health Sciences (DISSAL), University of Genoa, Largo R. Benzi 10, 16132, Genoa, Italy.
| | - Fabio Pastorino
- Laboratory of Experimental Therapy in Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | | | - Vanessa Cossu
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Anna Maria Orengo
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Patrizia Piccioli
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Laura Emionite
- Animal Facility, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Selene Capitanio
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nikola Yosifov
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Bruno
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Edoardo Lazzarini
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine & Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Mirco Ponzoni
- Laboratory of Experimental Therapy in Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine & Centre of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | - Anna Rubartelli
- Cell Biology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Ravera
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Silvia Morbelli
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Nuclear Medicine, Department of Health Sciences (DISSAL), University of Genoa, Largo R. Benzi 10, 16132, Genoa, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Nuclear Medicine, Department of Health Sciences (DISSAL), University of Genoa, Largo R. Benzi 10, 16132, Genoa, Italy
| | - Cecilia Marini
- Nuclear Medicine, Department of Health Sciences (DISSAL), University of Genoa, Largo R. Benzi 10, 16132, Genoa, Italy
- CNR Institute of Molecular Bioimaging and Physiology, Milan, Italy
| |
Collapse
|
31
|
Zeka K, Marrazzo P, Micucci M, Ruparelia KC, Arroo RRJ, Macchiarelli G, Annarita Nottola S, Continenza MA, Chiarini A, Angeloni C, Hrelia S, Budriesi R. Activity of Antioxidants from Crocus sativus L. Petals: Potential Preventive Effects towards Cardiovascular System. Antioxidants (Basel) 2020; 9:antiox9111102. [PMID: 33182461 PMCID: PMC7697793 DOI: 10.3390/antiox9111102] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 12/17/2022] Open
Abstract
The petals of the saffron crocus (Crocus sativus L.) are considered a waste material in saffron production, but may be a sustainable source of natural biologically active substances of nutraceutical interest. The aim of this work was to study the cardiovascular effects of kaempferol and crocin extracted from saffron petals. The antiarrhythmic, inotropic, and chronotropic effects of saffron petal extract (SPE), kaempferol, and crocin were evaluated through in vitro biological assays. The antioxidant activity of kaempferol and crocin was investigated through the 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) assay using rat cardiomyoblast cell line H9c2. The MTT assay was applied to assess the effects of kaempferol and crocin on cell viability. SPE showed weak negative inotropic and chronotropic intrinsic activities but a significant intrinsic activity on smooth muscle with a potency on the ileum greater than on the aorta: EC50 = 0.66 mg/mL versus EC50 = 1.45 mg/mL. Kaempferol and crocin showed a selective negative inotropic activity. In addition, kaempferol decreased the contraction induced by KCl (80 mM) in guinea pig aortic and ileal strips, while crocin had no effect. Furthermore, following oxidative stress, both crocin and kaempferol decreased intracellular ROS formation and increased cell viability in a concentration-dependent manner. The results indicate that SPE, a by-product of saffron cultivation, may represent a good source of phytochemicals with a potential application in the prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Keti Zeka
- Department of Haematology, Cambridge Biomedical Campus, University of Cambridge, Long Road, Cambridge CB2 0PT, UK;
| | - Pasquale Marrazzo
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (P.M.); (S.H.)
| | - Matteo Micucci
- Nutraceutical Lab, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (A.C.); (R.B.)
- Correspondence:
| | - Ketan C. Ruparelia
- Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (K.C.R.); (R.R.J.A.)
| | - Randolph R. J. Arroo
- Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK; (K.C.R.); (R.R.J.A.)
| | - Guido Macchiarelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy; (G.M.); (M.A.C.)
| | - Stefania Annarita Nottola
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, La Sapienza University of Rome, 00161 Rome, Italy;
| | - Maria Adelaide Continenza
- Department of Life, Health and Environmental Sciences, University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy; (G.M.); (M.A.C.)
| | - Alberto Chiarini
- Nutraceutical Lab, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (A.C.); (R.B.)
| | - Cristina Angeloni
- School of Pharmacy, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino (MC), Italy;
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy; (P.M.); (S.H.)
| | - Roberta Budriesi
- Nutraceutical Lab, Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy; (A.C.); (R.B.)
| |
Collapse
|
32
|
Czepas J, Matczak K, Koceva-Chyła A, Grobelski B, Jóźwiak Z, Gwoździński K. Doxyl Nitroxide Spin Probes Can Modify Toxicity of Doxorubicin towards Fibroblast Cells. Molecules 2020; 25:E5138. [PMID: 33158261 PMCID: PMC7663118 DOI: 10.3390/molecules25215138] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022] Open
Abstract
The biological properties of doxyl stearate nitroxides (DSs): 5-DS, Met-12-DS, and 16-DS, commonly used as spin probes, have not been explored in much detail so far. Furthermore, the influence of DSs on the cellular changes induced by the anticancer drug doxorubicin (DOX) has not yet been investigated. Therefore, we examined the cytotoxicity of DSs and their ability to induce cell death and to influence on fluidity and lipid peroxidation (LPO) in the plasma membrane of immortalised B14 fibroblasts, used as a model neoplastic cells, susceptible to DOX-induced changes. The influence of DSs on DOX toxicity was also investigated and compared with that of a natural reference antioxidant α-Tocopherol. By employing the trypan blue exclusion test and double fluorescent staining, we found a significant level of cytotoxicity for DSs and showed that their ability to induce apoptosis and modify plasma membrane fluidity (measured fluorimetrically) is more potent than for α-Tocopherol. The most cytotoxic nitroxide was 5-DS. The electron paramagnetic resonance (EPR) measurements revealed that 5-DS was reduced in B14 cells at the fastest and Met-12-DS at the slowest rate. In the presence of DOX, DSs were reduced slower than alone. The investigated compounds, administered with DOX, enhanced DOX-induced cell death and demonstrated concentration-dependent biphasic influence on membrane fluidity. A-Tocopherol showed weaker effects than DSs, regardless the mode of its application-alone or with DOX. High concentrations of α-Tocopherol and DSs decreased DOX-induced LPO. Substantial cytotoxicity of the DSs suggests that they should be used more carefully in the investigations performed on sensitive cells. Enhancement of DOX toxicity by DSs showed their potential to act as chemosensitizers of cancer cells to anthracycline chemotherapy.
Collapse
Affiliation(s)
- Jan Czepas
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (B.G.); (K.G.)
| | - Karolina Matczak
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (Z.J.)
| | - Aneta Koceva-Chyła
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (Z.J.)
| | - Bartłomiej Grobelski
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (B.G.); (K.G.)
| | - Zofia Jóźwiak
- Department of Medical Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (K.M.); (A.K.-C.); (Z.J.)
| | - Krzysztof Gwoździński
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Łódź, 141/143 Pomorska st., 90-236 Łódź, Poland; (B.G.); (K.G.)
| |
Collapse
|
33
|
Upadhyay S, Mantha AK, Dhiman M. Glycyrrhiza glabra (Licorice) root extract attenuates doxorubicin-induced cardiotoxicity via alleviating oxidative stress and stabilising the cardiac health in H9c2 cardiomyocytes. JOURNAL OF ETHNOPHARMACOLOGY 2020; 258:112690. [PMID: 32105749 DOI: 10.1016/j.jep.2020.112690] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 05/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Doxorubicin (DOX) is an effective anti-neoplastic drug, however; it has downside effects on cardiac health and other vital organs. The herbal remedies used in day to day life may have a beneficial effect without disturbing the health of the vital organs. Glycyrrhiza glabra L. is a ligneous perennial shrub belonging to Leguminosae/Fabaceae/Papilionaceae family growing in Mediterranean region and Asia and widespread in Turkey, Italy, Spain, Russia, Syria, Iran, China, India and Israel. Commonly known as mulaithi in north India, G. glabra has glycyrrhizin, glycyrrhetic acid, isoliquiritin, isoflavones, etc., which have been reported for several pharmacological activities such as anti-demulcent, anti-ulcer, anti-cancer, anti-inflammatory and anti-diabetic. AIM OF THE STUDY The objective of the present study is to investigate the interaction between the molecular factors like PPAR-α/γ and SIRT-1 during cardiac failure arbitrated by DOX under in vitro conditions and role of Glycyrrhiza glabra (Gg) root extract in alleviating these affects. MATERIALS AND METHODS In the present study, we have examined the DOX induced responses in H9c2 cardiomyocytes and investigated the role of phytochemical Glycyrrhiza glabra in modulating these affects. MTT assay was done to evaluate the cell viability, Reactive Oxygen Species (ROS)/Reactive Nitrogen Species (RNS) levels, mitochondrial ROS, mitochondrial membrane potential was estimated using fluorescent probes. The oxidative stress in terms of protein carbonylation, lipid peroxidation and DNA damage was detected via spectrophotometric methods and immune-fluorescence imaging. The cardiac markers and interaction between SIRT-1 and PPAR-α/γ was measured using Real-Time PCR, Western blotting and Co-immunoprecipitation based studies. RESULTS The Glycyrrhiza glabra (Gg) extracts maintained the membrane integrity and improved the lipid homeostasis and stabilized cytoskeletal element actin. Gg phytoextracts attenuated aggravated ROS level, repaired the antioxidant status and consequently, assisted in repairing the DNA damage and mitochondrial function. Further, the expression of hypertrophic markers in the DOX treated cardiomyocytes reconciled the expression factors both at the transcriptional and translational levels after Gg treatment. SIRT-1 mediated pathway and its downstream activator PPARs are significant in maintaining the cellular functions. It was observed that the Gg extract allows regaining the nuclear SIRT-1 and PPAR-γ level which was otherwise reduced with DOX treatment in H9c2 cardiomyocytes. The co-immunoprecipitation (Co-IP) documented that SIRT-1 interacts with PPAR-α in the untreated control H9c2 cardiomyocytes whereas DOX treatment interferes and diminishes this interaction however the Gg treatment maintains this interaction. Knocking down SIRT-1 also downregulated expression of PPAR-α and PPAR-γ in DOX treated cells and Gg treatment was able to enhance the expression of PPAR-α and PPAR-γ in SIRT-1 knocked down cardiomyocytes. CONCLUSIONS The antioxidant property of Gg defend the cardiac cells against the DOX induced toxicity via; 1) reducing the oxidative stress, 2) maintaining the mitochondrial functions, 3) regulating lipid homeostasis and cardiac metabolism through SIRT-1 pathway, and 4) conserving the cardiac hypertrophy and hence preserving the cardiomyocytes health. Therefore, Gg can be recommended as a healthy supplement with DOX towards cancer therapeutics associated cardiotoxicity.
Collapse
Affiliation(s)
- Shishir Upadhyay
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Anil Kumar Mantha
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151001, Punjab, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, 151001, Punjab, India.
| |
Collapse
|
34
|
Studneva IM, Veselova OM, Bahtin AA, Konovalova GG, Lankin VZ, Pisarenko OI. The Mechanisms of Cardiac Protection Using a Synthetic Agonist of Galanin Receptors during Chronic Administration of Doxorubicin. Acta Naturae 2020; 12:89-98. [PMID: 32477603 PMCID: PMC7245963 DOI: 10.32607/actanaturae.10945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The use of the anticancer drug doxorubicin (Dox) is limited by its cardiotoxic
effect. The aim of this work was to study the effect of a new synthetic agonist
of the galanin receptor GalR1-3 [βAla14, His15]-galanine (2–15) (G)
on the metabolism, antioxidant enzyme activity, and cardiac function in rats
with cardiomyopathy (CM) caused by chronic administration of Dox.
Coadministration of peptide G and Dox significantly increased the fractional
shortening (FS) and ejection fraction (EF) by an average of 30 ± 4%
compared with the indices in the Dox group. The reduced severity of cardiac
dysfunction under the action of G was accompanied by a 2.5-fold decrease in the
activity of creatine kinase-MB (CK-MB) in blood plasma. The protective
mechanism of the action of peptide G is caused by a reduced lipid peroxidation
(LP) that is due to the increased activity of Cu,Zn superoxide dismutase
(Cu,Zn-SOD) and glutathione peroxidase (GSH-Px) in the damaged heart.
Administration of peptide G significantly increased the adenine nucleotide pool
(ΣAH), ATP content, and the levels of phosphocreatine (PCr) and total
creatine (ΣCr) in the damaged myocardium. It also reduced lactate
accumulation relative to its content in the Dox group. The better energy supply
of cardiomyocytes after treatment with peptide G prevented the accumulation of
cytotoxic ammonia and disruption in the metabolism of the key myocardial amino
acids (glutamic acid (Glu), aspartic acid (Asp), and alanine (Ala)). Peptide G
significantly improved the morphological parameters of the heart in rats
treated with Dox. The results show promise in using peptide G to efficiently
correct functional, morphological, and metabolic damage to the heart caused by
anthracycline chemotherapy.
Collapse
Affiliation(s)
- I. M. Studneva
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| | - O. M. Veselova
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| | - A. A. Bahtin
- Research and Clinical Center of Otorhinolaryngology, Moscow, 123182 Russia
| | - G. G. Konovalova
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| | - V. Z. Lankin
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| | - O. I. Pisarenko
- National Medical Research Center for Cardiology, Moscow, 121552 Russia
| |
Collapse
|
35
|
Boussada M, Ali RB, Chahbi A, Abdelkarim M, Fradj MKB, Dziri C, Bokri K, Akacha AB, El May MV. A new Thiocyanoacetamide protects rat sperm cells from Doxorubicin-triggered cytotoxicity whereas Selenium shows low efficacy: In vitro approach. Toxicol In Vitro 2019; 61:104587. [PMID: 31271807 DOI: 10.1016/j.tiv.2019.06.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/29/2019] [Accepted: 06/29/2019] [Indexed: 11/26/2022]
Abstract
Doxorubicin (DOX) exhibits a wide-ranging spectrum of antitumor activities which maintain its clinical use despite its devastating impact on highly proliferating cells. The present work was designed to develop a new approach which aims to protect male germ cells from DOX cytotoxicity. Thus, an assessment of the protective potential of a new thioamide analog (thiocyanoacetamide; TA) compared to selenium (Se) was performed in rat sperms exposed to DOX in vitro. Oxygen consumption rate (OCR) was measured after exposure to three different doses (0.5, 1, 1.5 and 2 μM) of DOX, Se or TA, and the suitable concentrations were selected for further studies afterwards. Motility, OCR in a time-dependent manner, glucose extracellular concentration and lipid peroxidation (LPO) were measured. Fatty acid (FA) content was assessed by gas chromatography (GC-FID). Cell death, superoxide anion (O2-), mitochondrial membrane potential (MMP), and DNA damage were evaluated by flow cytometry. TA association with DOX increased OCR and glucose uptake, improved cell survival and decreased DNA damage. The co-administration of DOX with Se increased OCR, significantly prevented O2- overproduction, and decreased LPO. Collected data brought new insights regarding this transformed TA, which showed better efficiency than Se in reducing DOX cytotoxic stress in sperms.
Collapse
Affiliation(s)
- Marwa Boussada
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia.
| | - Ridha Ben Ali
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia; Unity of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Ahlem Chahbi
- Laboratory of Hematology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia.
| | - Mohamed Abdelkarim
- Laboratory of Hematology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Mohamed Kacem Ben Fradj
- UR05/08-08, LR99/ES/11, Department of Biochemistry, Rabta Hospital, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Chadli Dziri
- Unity of Experimental Medicine, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| | - Khouloud Bokri
- Laboratory of Organic Synthesis and Heterocyclic Chemistry Department, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092 Tunis, Tunisia
| | - Azaiez Ben Akacha
- Laboratory of Organic Synthesis and Heterocyclic Chemistry Department, Faculty of Sciences of Tunis, University of Tunis El Manar, 2092 Tunis, Tunisia
| | - Michèle Véronique El May
- UR17/ES/13 Laboratory of Histology and Embryology, Faculty of Medicine of Tunis, University of Tunis El Manar (UTM), Jabbari Jebel Lakhdar Street 15, 1007 Tunis, Tunisia
| |
Collapse
|
36
|
FDG-PET Imaging of Doxorubicin-Induced Cardiotoxicity: a New Window on an Old Problem. CURRENT CARDIOVASCULAR IMAGING REPORTS 2019. [DOI: 10.1007/s12410-019-9517-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
37
|
Studneva IM, Palkeeva ME, Veselova OM, Molokoedov AS, Lubimov RO, Ovchinnikov MV, Sidorova MV, Pisarenko OI. [Protective action of a modified fragment of galanine in rats with doxorubicin-induced heart failure]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 65:51-56. [PMID: 30816097 DOI: 10.18097/pbmc20196501051] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The use of the anticancer drug doxorubicin (Dox) is limited due to its cardiotoxic effect. Using the method of automatic solid-phase peptide synthesis, we obtained a synthetic agonist of galanin receptors GalR1-3 [RAla14, His15]-galanine (2-15) (G), exhibiting cardioprotective properties. It was purified by high performance liquid chromatography (HPLC). The homogeneity and structure of the peptide was confirmed by HPLC, 1H-NMR spectroscopy and mass spectroscopy. The purpose of this study was to study the effect of G on the metabolism and cardiac function of rats with chronic heart failure (CHF) caused by Dox. Experiments were performed using male Wistar rats weighing 280-300 g. The control group of animals (C) was intraperitoneally treated with saline for 8 weeks; the doxorubicin group (D) of rats was intraperitoneally treated with Doх; the group of Doх + peptide G (D+G) received intraperitoneally injections of Doх and subcutaneously injections of peptide G; the peptide G group (G) was subcutaneously treated with G. At the beginning and at the end of the study, the concentration of thiobarbituric acid reactive substances (TBARS) and the activity of creatine kinase-MB (CK-MB) were determined in blood plasma; the animals were weighed, and cardiac function was assessed using echocardiography. At the end of the experiments, the hearts were used for determination of metabolites and assessment of oxidative phosphorylation in mitochondria. After 8-week treatment, animals of group D were characterized by severe heart failure, the lack of weight gain and an increase in plasma TBARS concentration and CK-MB activity. These disorders were accompanied by a decrease in the content of myocardial high-energy phosphates, a reduction inmitochondrial respiratory parameters, accumulation of lactate and glucose in the heart, and disturbances in the metabolism of alanine and glutamic and aspartic acids. Coadministration of G and Dox prevented the increase in plasma CK-MB activity and significantly reduced the plasma TBARS concentration. At the end of the experiments animals of group D+G had higher myocardial energy state and the respiratory control index of mitochondria than animals of group D, there was a decrease in anaerobic glycolysis and no changes in the amino acid content compared to the control. The peptide G significantly improved the parameters of cardiac function and caused weight gain in animals of group D+G in comparison with these parameters in group D. The obtained results demonstrate the ability of a novel agonist of galanin receptors GalR1-3 to attenuate Dox-indiced cardiotoxicity.
Collapse
Affiliation(s)
- I M Studneva
- National Medical Research Center for Cardiology, Moscow, Russia
| | - M E Palkeeva
- National Medical Research Center for Cardiology, Moscow, Russia
| | - O M Veselova
- National Medical Research Center for Cardiology, Moscow, Russia
| | - A S Molokoedov
- National Medical Research Center for Cardiology, Moscow, Russia
| | - R O Lubimov
- National Medical Research Center for Cardiology, Moscow, Russia
| | - M V Ovchinnikov
- National Medical Research Center for Cardiology, Moscow, Russia
| | - M V Sidorova
- National Medical Research Center for Cardiology, Moscow, Russia
| | - O I Pisarenko
- National Medical Research Center for Cardiology, Moscow, Russia
| |
Collapse
|
38
|
Blockade of L-type Ca 2+ channel attenuates doxorubicin-induced cardiomyopathy via suppression of CaMKII-NF-κB pathway. Sci Rep 2019; 9:9850. [PMID: 31285514 PMCID: PMC6614470 DOI: 10.1038/s41598-019-46367-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 06/27/2019] [Indexed: 12/30/2022] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) and nuclear factor-kappa B (NF-κB) play crucial roles in pathogenesis of doxorubicin (DOX)-induced cardiomyopathy. Their activities are regulated by intracellular Ca2+. We hypothesized that blockade of L-type Ca2+ channel (LTCC) could attenuate DOX-induced cardiomyopathy by regulating CaMKII and NF-κB. DOX activated CaMKII and NF-κB through their phosphorylation and increased cleaved caspase 3 in cardiomyocytes. Pharmacological blockade or gene knockdown of LTCC by nifedipine or small interfering RNA, respectively, suppressed DOX-induced phosphorylation of CaMKII and NF-κB and apoptosis in cardiomyocytes, accompanied by decreasing intracellular Ca2+ concentration. Autocamtide 2-related inhibitory peptide (AIP), a selective CaMKII inhibitor, inhibited DOX-induced phosphorylation of NF-κB and cardiomyocyte apoptosis. Inhibition of NF-κB activity by ammonium pyrrolidinedithiocarbamate (PDTC) suppressed DOX-induced cardiomyocyte apoptosis. DOX-treatment (18 mg/kg via intravenous 3 injections over 1 week) increased phosphorylation of CaMKII and NF-κB in mouse hearts. Nifedipine (10 mg/kg/day) significantly suppressed DOX-induced phosphorylation of CaMKII and NF-κB and cardiomyocyte injury and apoptosis in mouse hearts. Moreover, it attenuated DOX-induced left ventricular dysfunction and dilatation. Our findings suggest that blockade of LTCC attenuates DOX-induced cardiomyocyte apoptosis via suppressing intracellular Ca2+ elevation and activation of CaMKII-NF-κB pathway. LTCC blockers might be potential therapeutic agents against DOX-induced cardiomyopathy.
Collapse
|
39
|
Coliva G, Duarte S, Pérez-Sala D, Fedorova M. Impact of inhibition of the autophagy-lysosomal pathway on biomolecules carbonylation and proteome regulation in rat cardiac cells. Redox Biol 2019; 23:101123. [PMID: 30737170 PMCID: PMC6859560 DOI: 10.1016/j.redox.2019.101123] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 01/13/2023] Open
Abstract
Cells employ multiple defence mechanisms to sustain a wide range of stress conditions associated with accumulation of modified self-biomolecules leading to lipo- and proteotoxicity. One of such mechanisms involves activation of the autophagy-lysosomal pathway for removal and degradation of modified lipids, proteins and even organelles. Biomolecules carbonylation, an irreversible oxidative modification, occurs in a variety of pathological conditions and is generally viewed as a marker of oxidative stress. Here, we used a model of rat primary cardiac cells to elucidate the role of autophagy-lysosomal pathway in the turnover of carbonylated biomolecules. Cells treated with inhibitors of autophagy-lysosomal degradation and primed with a short pulse of mild nitroxidative stress were studied using fluorescent microscopy and accumulation of carbonylated biomolecules in droplets- or vesicle-like structures was observed. Furthermore, systems-wide analysis of proteome regulation using relative label free quantification approach revealed the most significant alterations in cells treated with protease inhibitors. Interestingly, down-regulation of insulin signalling was among the most enriched pathway, as revealed by functional annotation of regulated proteins. Starvation induced autophagy promotes cellular carbonylation. Inhibition of autophagy-lysosomal flux leads to carbonyls accumulation. Cellular carbonyls coincide with Nile Red positive structures. Inhibition of autophagy-lysosomal flux induces proteome alterations. Impairment of autophagy-lysosomal flux results in changes in metabolic and nutrient sensing pathways.
Collapse
Affiliation(s)
- Giulia Coliva
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Deutscher Platz 5, 04103 Leipzig, Germany; Center for Biotechnology and Biomedicine, University of Leipzig, Deutscher Platz 5, 04103 Leipzig, Germany
| | - Sofia Duarte
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas, C.S.I.C., 28040 Madrid, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas, C.S.I.C., 28040 Madrid, Spain
| | - Maria Fedorova
- Institute of Bioanalytical Chemistry, Faculty of Chemistry and Mineralogy, University of Leipzig, Deutscher Platz 5, 04103 Leipzig, Germany; Center for Biotechnology and Biomedicine, University of Leipzig, Deutscher Platz 5, 04103 Leipzig, Germany.
| |
Collapse
|
40
|
Arroyo-Crespo JJ, Armiñán A, Charbonnier D, Balzano-Nogueira L, Huertas-López F, Martí C, Tarazona S, Forteza J, Conesa A, Vicent MJ. Tumor microenvironment-targeted poly-L-glutamic acid-based combination conjugate for enhanced triple negative breast cancer treatment. Biomaterials 2018; 186:8-21. [DOI: 10.1016/j.biomaterials.2018.09.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 09/11/2018] [Accepted: 09/14/2018] [Indexed: 12/26/2022]
|
41
|
Andrade N, Silva C, Martel F. The effect of oxidative stress upon intestinal sugar transport: an in vitro study using human intestinal epithelial (Caco-2) cells. Toxicol Res (Camb) 2018; 7:1236-1246. [PMID: 30542607 PMCID: PMC6243649 DOI: 10.1039/c8tx00183a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/10/2018] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of various gastrointestinal diseases, including gastrointestinal cancers and inflammatory bowel disease, is associated with increased oxidative stress levels. We aimed to investigate the effect of oxidative stress induced by tert-butylhydroperoxide (TBH) on the uptake of 3H-deoxy-d-glucose (3H-DG) and 14C-fructose by the human intestinal Caco-2 cell line. TBH (500 μM; 24 h) increased lipid peroxidation (TBARS) levels and was not cytotoxic. TBH (500 μM; 24 h) increased uptake of both low (SGLT1-mediated) and high concentrations (SGLT1- and GLUT2-mediated) of 3H-DG, but did not affect absorption of 14C-fructose (GLUT2- and GLUT5-mediated). The polyphenol chrysin abolished the increase in TBARS levels and the increase in uptake of both low and high concentrations of 3H-DG induced by TBH. On the other hand, TBH blocked the inhibitory effect of chrysin on 14C-fructose uptake. 3H-DG uptake, but not 14C-fructose uptake, was sensitive to sweet taste receptor (STRs) inhibition (with lactisole). The inhibitory effect of lactisole in relation to uptake of 3H-DG (10 nM) (SGLT1-mediated), but not in relation to uptake of 3H-DG (50 mM) (SGLT1- and GLUT2-mediated), was abolished in the presence of TBH. So, these results show that the stimulatory effect of STRs on SGLT1-mediated transport is dependent on oxidative stress levels. In conclusion, this work shows that uptake of both 3H-DG and 14C-fructose is sensitive to oxidative stress levels. Moreover, it suggests that the three distinct transporters involved in the intestinal absorption of glucose and fructose (SGLT1, GLUT2 and GLUT5) have different sensitivities to oxidative stress levels, SGLT1 being the most sensitive and GLUT5 the least.
Collapse
Affiliation(s)
- Nelson Andrade
- Department of Biomedicine - Unit of Biochemistry , Faculty of Medicine of Porto , University of Porto , Porto , Portugal .
- Instituto de Investigação e Inovação em Saúde (I3S) , University of Porto , Porto , Portugal
| | - Cláudia Silva
- Department of Biomedicine - Unit of Biochemistry , Faculty of Medicine of Porto , University of Porto , Porto , Portugal .
- Instituto de Investigação e Inovação em Saúde (I3S) , University of Porto , Porto , Portugal
| | - Fátima Martel
- Department of Biomedicine - Unit of Biochemistry , Faculty of Medicine of Porto , University of Porto , Porto , Portugal .
- Instituto de Investigação e Inovação em Saúde (I3S) , University of Porto , Porto , Portugal
| |
Collapse
|
42
|
Sarocchi M, Bauckneht M, Arboscello E, Capitanio S, Marini C, Morbelli S, Miglino M, Congiu AG, Ghigliotti G, Balbi M, Brunelli C, Sambuceti G, Ameri P, Spallarossa P. An increase in myocardial 18-fluorodeoxyglucose uptake is associated with left ventricular ejection fraction decline in Hodgkin lymphoma patients treated with anthracycline. J Transl Med 2018; 16:295. [PMID: 30359253 PMCID: PMC6202821 DOI: 10.1186/s12967-018-1670-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/17/2018] [Indexed: 12/19/2022] Open
Abstract
Background Doxorubicin (DOX)-based chemotherapy for Hodgkin lymphoma (HL) yields excellent disease-free survival, but poses a substantial risk of subsequent left ventricular (LV) dysfunction and heart failure, typically with delayed onset. At the cellular level, this cardiotoxicity includes deranged cardiac glucose metabolism. Methods By reviewing the hospital records from January 2008 through December 2016, we selected HL patients meeting the following criteria: ≥ 18 year-old; first-line DOX-containing chemotherapy; no diabetes and apparent cardiovascular disease; 18-fluoro-deoxyglucose positron emission tomography (18FDG-PET) scans before treatment (PETSTAGING), after 2 cycles (PETINTERIM) and at the end of treatment (PETEOT); at least one echocardiography ≥ 6 months after chemotherapy completion (ECHOPOST). We then evaluated the changes in LV 18FDG standardized uptake values (SUV) during the course of DOX therapy, and the relationship between LV-SUV and LV ejection fraction (LVEF), as calculated from the LV diameters in the echocardiography reports with the Teicholz formula. Results Forty-three patients (35 ± 13 year-old, 58% males) were included in the study, with 26 (60%) also having a baseline echocardiography available (ECHOPRE). LV-SUV gradually increased from PETSTAGING (log-transformed mean 0.20 ± 0.27) to PETINTERIM (0.27 ± 0.35) to PETEOT (0.30 ± 0.41; P for trend < 0.001). ECHOPOST was performed 22 ± 17 months after DOX chemotherapy. Mean LVEF was normal (68.8 ± 10.3%) and only three subjects (7%) faced a drop below the upper normal limit of 53%. However, when patients were categorized by median LV-SUV, LVEF at ECHOPOST resulted significantly lower in those with LV-SUV above than below the median value at both PETINTERIM (65.5 ± 11.8% vs. 71.9 ± 7.8%, P = 0.04) and PETEOT (65.6 ± 12.2% vs. 72.2 ± 7.0%, P = 0.04). This was also the case when only patients with ECHOPRE and ECHOPOST were considered (LVEF at ECHOPOST 64.7 ± 8.9% vs. 73.4 ± 7.6%, P = 0.01 and 64.6 ± 9.3% vs. 73.5 ± 7.0%, P = 0.01 for those with LV-SUV above vs. below the median at PETINTERIM and PETEOT, respectively). Furthermore, the difference between LVEF at ECHOPRE and ECHOPOST was inversely correlated with LV-SUV at PETEOT (P < 0.01, R2 = − 0.30). Conclusions DOX-containing chemotherapy causes an increase in cardiac 18FDG uptake, which is associated with a decline in LVEF. Future studies are warranted to understand the molecular basis and the potential clinical implications of this observation.
Collapse
Affiliation(s)
- Matteo Sarocchi
- Cardiovascular Diseases Unit, IRCCS Ospedale Policlinico San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy.,Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Matteo Bauckneht
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Health Sciences, University of Genova, Genoa, Italy
| | - Eleonora Arboscello
- Emergency Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Selene Capitanio
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Cecilia Marini
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,CNR Institute of Molecular Bioimaging and Physiology, Milan, Italy
| | - Silvia Morbelli
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Health Sciences, University of Genova, Genoa, Italy
| | - Maurizio Miglino
- Department of Internal Medicine, University of Genova, Genoa, Italy.,Haematology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Giorgio Ghigliotti
- Cardiovascular Diseases Unit, IRCCS Ospedale Policlinico San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy.,Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Manrico Balbi
- Cardiovascular Diseases Unit, IRCCS Ospedale Policlinico San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy.,Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Claudio Brunelli
- Cardiovascular Diseases Unit, IRCCS Ospedale Policlinico San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy.,Department of Internal Medicine, University of Genova, Genoa, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,CNR Institute of Molecular Bioimaging and Physiology, Milan, Italy
| | - Pietro Ameri
- Cardiovascular Diseases Unit, IRCCS Ospedale Policlinico San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy. .,Department of Internal Medicine & Center of Excellence for Biomedical Research, University of Genova, Genoa, Italy.
| | - Paolo Spallarossa
- Cardiovascular Diseases Unit, IRCCS Ospedale Policlinico San Martino, IRCCS Italian Cardiovascular Network, Genova, Italy
| |
Collapse
|
43
|
Polyakov N, Leshina T, Fedenok L, Slepneva I, Kirilyuk I, Furso J, Olchawa M, Sarna T, Elas M, Bilkis I, Weiner L. Redox-Active Quinone Chelators: Properties, Mechanisms of Action, Cell Delivery, and Cell Toxicity. Antioxid Redox Signal 2018; 28:1394-1403. [PMID: 29161882 DOI: 10.1089/ars.2017.7406] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
SIGNIFICANCE Chemotherapy is currently the principal method for treating many malignancies. Thus, the development of improved antitumor drugs with enhanced efficacy and selectivity remains a high priority. Recent Advances: Anthracycline antibiotics (AAs), for example, doxorubicin, daunomycin, and mitomycin C, belong to an important family of antitumor agents widely used in chemotherapy. These compounds are all quinones. They are, thus, capable of being reduced by appropriate chemicals or reductases. One of their important properties is that under aerobic conditions their reduced forms undergo oxidation, with concomitant generation of reactive oxygen species (ROS), namely, superoxide anion radicals, hydrogen peroxide, and hydroxyl radicals. The presence of metal ions is essential for the generation of ROS by AAs in biological systems. CRITICAL ISSUES A fundamental shortcoming of the AAs is their high cardiotoxicity. We have proposed, and experimentally realized, a new type of quinones that is capable of coordinating metal ions. We have demonstrated in vitro that they can be reduced by electron transfer chains and glutathione with concomitant generation of ROS. They can also produce ROS under photo-excitation. The mechanisms of these reactions have been characterized by using nuclear magnetic resonance and electron paramagnetic resonance. FUTURE DIRECTIONS To enhance their therapeutic effectiveness, and decrease cardiotoxicity and other side effects, we intend to conjugate the quinone chelators with monoclonal antibodies and peptide hormones that are specifically targeted to receptors on the cancer cell surface. Some such candidates have already been synthesized. An alternative approach for delivery of our compounds involves the use of specific peptide-based nanoparticles. In addition, our novel approach for treating malignancies is also suitable for photodynamic therapy. Antioxid. Redox Signal. 28, 1394-1403.
Collapse
Affiliation(s)
- Nikolay Polyakov
- 1 Institute of Chemical Kinetics and Combustion , Novosibirsk, Russia
| | - Tatyana Leshina
- 1 Institute of Chemical Kinetics and Combustion , Novosibirsk, Russia
| | - Lidiya Fedenok
- 1 Institute of Chemical Kinetics and Combustion , Novosibirsk, Russia
| | - Irina Slepneva
- 1 Institute of Chemical Kinetics and Combustion , Novosibirsk, Russia
| | - Igor Kirilyuk
- 2 Laboratory of Nitrogen Compounds, Novosibirsk Institute of Organic Chemistry , Novosibirsk, Russia
| | - Justyna Furso
- 3 Department of Biophysics, Jagiellonian University , Kraków, Poland
| | - Magdalena Olchawa
- 3 Department of Biophysics, Jagiellonian University , Kraków, Poland
| | - Tadeusz Sarna
- 3 Department of Biophysics, Jagiellonian University , Kraków, Poland
| | - Martyna Elas
- 3 Department of Biophysics, Jagiellonian University , Kraków, Poland
| | - Itzhak Bilkis
- 4 Institute of Biochemistry, Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem , Rehovot, Israel
| | - Lev Weiner
- 5 Department of Neurobiology, Faculty of Biology, Weizmann Institute of Science , Rehovot, Israel
| |
Collapse
|
44
|
Prata C, Zambonin L, Rizzo B, Maraldi T, Angeloni C, Vieceli Dalla Sega F, Fiorentini D, Hrelia S. Glycosides from Stevia rebaudiana Bertoni Possess Insulin-Mimetic and Antioxidant Activities in Rat Cardiac Fibroblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3724545. [PMID: 28947927 PMCID: PMC5602648 DOI: 10.1155/2017/3724545] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/11/2017] [Indexed: 12/29/2022]
Abstract
Stevia rebaudiana Bertoni is a shrub having a high content of sweet diterpenoid glycosides in its leaves, mainly stevioside and rebaudioside A, which are used as noncaloric, natural sweeteners. The aim of this study was to deepen the knowledge about the insulin-mimetic effect exerted by four different mixtures of steviol glycosides, rich in stevioside and rebaudioside A, in neonatal rat cardiac fibroblasts. The potential antioxidant activity of these steviol glycosides was also assessed, as oxidative stress is associated with diabetes. Likewise the insulin effect, steviol glycosides caused an increase in glucose uptake into rat fibroblasts by activating the PI3K/Akt pathway, thus inducing Glut4 translocation to the plasma membrane. The presence of S961, an insulin antagonist, completely abolished these effects, allowing to hypothesize that steviol glycosides could act as ligands of the same receptor engaged by insulin. Moreover, steviol glycosides counteracted oxidative stress by increasing reduced glutathione intracellular levels and upregulating expression and activity of the two antioxidant enzymes superoxide dismutase and catalase. The present work unravels the insulin-mimetic effect and the antioxidant property exerted by steviol glycosides, suggesting their potential beneficial role in the cotreatment of diabetes and in health maintenance.
Collapse
Affiliation(s)
- Cecilia Prata
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio, No. 48, 40126 Bologna, Italy
| | - Laura Zambonin
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio, No. 48, 40126 Bologna, Italy
| | - Benedetta Rizzo
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, Corso d'Augusto, No. 237, 47921 Rimini, Italy
| | - Tullia Maraldi
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Policlinico, Via del Pozzo, No. 71, 41124 Modena, Italy
| | - Cristina Angeloni
- School of Pharmacy, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy
| | | | - Diana Fiorentini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Via Irnerio, No. 48, 40126 Bologna, Italy
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, Corso d'Augusto, No. 237, 47921 Rimini, Italy
| |
Collapse
|
45
|
Dolinsky VW. The role of sirtuins in mitochondrial function and doxorubicin-induced cardiac dysfunction. Biol Chem 2017; 398:955-974. [PMID: 28253192 DOI: 10.1515/hsz-2016-0316] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/11/2017] [Indexed: 01/15/2023]
Abstract
Anthracycline chemotherapeutics such as doxorubicin continue to be important treatments for many cancers. Through improved screening and therapy, more patients are surviving and living longer after the diagnosis of their cancer. However, anthracyclines are associated with both short- and long-term cardiotoxic effects. Doxorubicin-induced mitochondrial dysfunction is a central mechanism in the cardiotoxic effects of doxorubicin that contributes to impaired cardiac energy levels, increased reactive oxygen species production, cardiomyocyte apoptosis and the decline in cardiac function. Sirtuins are protein deacetylases that are activated by low energy levels and stimulate energy production through their activation of transcription factors and enzymatic regulators of cardiac energy metabolism. In addition, sirtuins activate oxidative stress resistance pathways. SIRT1 and SIRT3 are expressed at high levels in the cardiomyocyte. This review examines the function of sirtuins in the regulation of cardiac mitochondrial function, with a focus on their role in heart failure and an emphasis on their effects on doxorubicin-induced cardiotoxicity. We discuss the potential for sirtuin activation in combination with anthracycline chemotherapy in order to mitigate its cardiotoxic side-effects without reducing the antineoplastic activity of anthracyclines.
Collapse
|
46
|
Bauckneht M, Ferrarazzo G, Fiz F, Morbelli S, Sarocchi M, Pastorino F, Ghidella A, Pomposelli E, Miglino M, Ameri P, Emionite L, Ticconi F, Arboscello E, Buschiazzo A, Massimelli EA, Fiordoro S, Borra A, Cossu V, Bozzano A, Ibatici A, Ponzoni M, Spallarossa P, Gallamini A, Bruzzi P, Sambuceti G, Marini C. Doxorubicin Effect on Myocardial Metabolism as a Prerequisite for Subsequent Development of Cardiac Toxicity: A Translational 18F-FDG PET/CT Observation. J Nucl Med 2017. [PMID: 28646013 DOI: 10.2967/jnumed.117.191122] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The present translational study aimed to verify whether serial 18F-FDG PET/CT predicts doxorubicin cardiotoxicity. Methods: Fifteen athymic mice were treated intravenously with saline (n = 5) or with 5 or 7.5 mg of doxorubicin per kilogram (n = 5 each) and underwent dynamic small-animal PET beforehand and afterward to estimate left ventricular (LV) metabolic rate of glucose (MRGlu). Thereafter, we retrospectively identified 69 patients who had been successfully treated with a regimen of doxorubicin, bleomycin, vinblastine, and dacarbazine for Hodgkin disease (HD) and had undergone 4 consecutive 18F-FDG PET/CT scans. Volumes of interest were drawn on LV myocardium to quantify mean SUV. All patients were subsequently interviewed by telephone (median follow-up, 30 mo); 36 of them agreed to undergo electrocardiography and transthoracic echocardiography. Results: In mice, LV MRGlu was 17.9 ± 4.4 nmol × min-1 × g-1 at baseline. Doxorubicin selectively and dose-dependently increased this value in the standard-dose (27.9 ± 9 nmol × min-1 × g-1, P < 0.05 vs. controls) and high-dose subgroups (37.2 ± 7.8 nmol × min-1 × g-1, P < 0.01 vs. controls, P < 0.05 vs. standard-dose). In HD patients, LV SUV showed a progressive increase during doxorubicin treatment that persisted at follow-up. New-onset cardiac abnormalities appeared in 11 of 36 patients (31%). In these subjects, pretherapy LV SUV was markedly lower with respect to the remaining patients (1.53 ± 0.9 vs. 3.34 ± 2.54, respectively, P < 0.01). Multivariate analysis confirmed the predictive value of baseline LV SUV for subsequent cardiac abnormalities. Conclusion: Doxorubicin dose-dependently increases LV MRGlu, particularly in the presence of low baseline 18F-FDG uptake. These results imply that low myocardial 18F-FDG uptake before the initiation of doxorubicin chemotherapy in HD patients may predict the development of chemotherapy-induced cardiotoxicity, suggesting that prospective clinical trials are warranted to test this hypothesis.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Giulia Ferrarazzo
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Francesco Fiz
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy.,Nuclear Medicine Unit, Department of Radiology, Tübingen, Germany
| | - Silvia Morbelli
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Matteo Sarocchi
- Clinic of Cardiovascular Diseases, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Fabio Pastorino
- Unit of Experimental Therapy in Oncology, IRCCS Gaslini, Genoa, Italy
| | - Alberto Ghidella
- Clinic of Cardiovascular Diseases, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Elena Pomposelli
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Maurizio Miglino
- Haematology Clinic, University of Genoa, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Pietro Ameri
- Clinic of Cardiovascular Diseases, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Laura Emionite
- Animal Facility, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Flavia Ticconi
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | | | - Ambra Buschiazzo
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | | | - Salvatore Fiordoro
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Anna Borra
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Vanessa Cossu
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Annalisa Bozzano
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Adalberto Ibatici
- Haematology Clinic, University of Genoa, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Mirco Ponzoni
- Unit of Experimental Therapy in Oncology, IRCCS Gaslini, Genoa, Italy
| | - Paolo Spallarossa
- Clinic of Cardiovascular Diseases, IRCCS-AOU San Martino-IST, Genoa, Italy
| | - Andrea Gallamini
- Department of Research, Innovation, and Statistics, Lacassagne Cancer Centre, Nice, France
| | - Paolo Bruzzi
- Epidemiology Unit, IRCCS-AOU San Martino-IST, Genoa, Italy; and
| | - Gianmario Sambuceti
- Nuclear Medicine, IRCCS-AOU San Martino-IST and University of Genoa, Genoa, Italy
| | - Cecilia Marini
- CNR Institute of Bioimaging and Molecular Physiology, Section of Genoa, Milan, Italy
| |
Collapse
|
47
|
Farhane Z, Bonnier F, Maher MA, Bryant J, Casey A, Byrne HJ. Differentiating responses of lung cancer cell lines to Doxorubicin exposure: in vitro Raman micro spectroscopy, oxidative stress and bcl-2 protein expression. JOURNAL OF BIOPHOTONICS 2017; 10:151-165. [PMID: 27088439 DOI: 10.1002/jbio.201600019] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 02/23/2016] [Accepted: 03/14/2016] [Indexed: 06/05/2023]
Abstract
The potential of Raman micro spectroscopy as an in vitro, non-invasive tool for clinical applications has been demonstrated in recent years, specifically for cancer research. To further illustrate its potential as a high content and label free technique, it is important to show its capability to elucidate drug mechanisms of action and cellular resistances. In this study, cytotoxicity assays were employed to establish the toxicity profiles for 24 hr exposure of lung cancer cell lines, A549 and Calu-1, to the commercially available drug, doxorubicin (DOX). Raman spectroscopy, coupled with Confocal Laser Scanning Microscopy and Flow Cytometry, was used to track the DOX mechanism of action, at a subcellular level, and to study the mechanisms of cellular resistance to DOX. Biomarkers related to the drug mechanism of action and cellular resistance to apoptosis, namely reactive oxygen species (ROS) and bcl-2 protein expression, respectively, were also measured and correlated to Raman spectral profiles. Calu-1 cells are shown to exhibit spectroscopic signatures of both direct DNA damage due to intercalation in the nucleus and indirect damage due to oxidative stress in the cytoplasm, whereas the A549 cell line only exhibits signatures of the former mechanism of action. PCA of nucleolar, nuclear and cytoplasmic regions of A549 and Calu-1 with corresponding loadings of PC1 and PC2.
Collapse
Affiliation(s)
- Zeineb Farhane
- FOCAS Research Institute, Dublin Institute of Technology, Kevin Street, Dublin, 8, Ireland
- School of Physics, Dublin Institute of Technology, Kevin Street, Dublin, 8, Ireland
| | - Franck Bonnier
- Université François-Rabelais de Tours, Faculty of Pharmacy, EA 6295 Nanomédicaments et Nanosondes, 31 avenue Monge, 37200, Tours, France
| | - Marcus Alexander Maher
- FOCAS Research Institute, Dublin Institute of Technology, Kevin Street, Dublin, 8, Ireland
- School of Physics, Dublin Institute of Technology, Kevin Street, Dublin, 8, Ireland
| | - Jane Bryant
- FOCAS Research Institute, Dublin Institute of Technology, Kevin Street, Dublin, 8, Ireland
| | - Alan Casey
- FOCAS Research Institute, Dublin Institute of Technology, Kevin Street, Dublin, 8, Ireland
| | - Hugh James Byrne
- FOCAS Research Institute, Dublin Institute of Technology, Kevin Street, Dublin, 8, Ireland
| |
Collapse
|
48
|
Kankeu C, Clarke K, Passante E, Huber HJ. Doxorubicin-induced chronic dilated cardiomyopathy-the apoptosis hypothesis revisited. J Mol Med (Berl) 2016; 95:239-248. [PMID: 27933370 DOI: 10.1007/s00109-016-1494-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/17/2016] [Accepted: 11/25/2016] [Indexed: 01/08/2023]
Abstract
The chemotherapeutic agent doxorubicin (DOX) has significantly increased survival rates of pediatric and adult cancer patients. However, 10% of pediatric cancer survivors will 10-20 years later develop severe dilated cardiomyopathy (DCM), whereby the exact molecular mechanisms of disease progression after this long latency time remain puzzling. We here revisit the hypothesis that elevated apoptosis signaling or its increased likelihood after DOX exposure can lead to an impairment of cardiac function and cause a cardiac dilation. Based on recent literature evidence, we first argue why a dilated phenotype can occur when little apoptosis is detected. We then review findings suggesting that mature cardiomyocytes are protected against DOX-induced apoptosis downstream, but not upstream of mitochondrial outer membrane permeabilisation (MOMP). This lack of MOMP induction is proposed to alter the metabolic phenotype, induce hypertrophic remodeling, and lead to functional cardiac impairment even in the absence of cardiomyocyte apoptosis. We discuss findings that DOX exposure can lead to increased sensitivity to further cardiomyocyte apoptosis, which may cause a gradual loss in cardiomyocytes over time and a compensatory hypertrophic remodeling after treatment, potentially explaining the long lag time in disease onset. We finally note similarities between DOX-exposed cardiomyocytes and apoptosis-primed cancer cells and propose computational system biology as a tool to predict patient individual DOX doses. In conclusion, combining recent findings in rodent hearts and cardiomyocytes exposed to DOX with insights from apoptosis signal transduction allowed us to obtain a molecularly deeper insight in this delayed and still enigmatic pathology of DCM.
Collapse
Affiliation(s)
- Cynthia Kankeu
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Kylie Clarke
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Egle Passante
- School of Pharmacy and Biomedical Sciences, Univ. of Central Lancashire, Preston, UK
| | - Heinrich J Huber
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium. .,Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St Stephens Green, Dublin 2, Ireland.
| |
Collapse
|
49
|
Chaiswing L, Cole MP, St Clair DK, Ittarat W, Szweda LI, Oberley TD. Oxidative Damage Precedes Nitrative Damage in Adriamycin-Induced Cardiac Mitochondrial Injury. Toxicol Pathol 2016; 32:536-47. [PMID: 15605432 DOI: 10.1080/01926230490502601] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The purpose of the present study was to determine if elevated reactive oxygen (ROS)/nitrogen species (RNS) reported to be present in adriamycin (ADR)-induced cardiotoxicity actually resulted in cardiomyocyte oxidative/nitrative damage, and to quantitatively determine the time course and subcellular localization of these postulated damage products using an in vivo approach. B6C3 mice were treated with a single dose of 20 mg/kg ADR. Ultrastructural damage and levels of 4-hydroxy-2-nonenal (4HNE)-protein adducts and 3-nitrotyrosine (3NT) were analyzed. Quantitative ultrastructural damage using computerized image techniques showed cardiomyocyte injury as early as 3 hours, with mitochondria being the most extensively and progressively injured subcellular organelle. Analysis of 4HNE protein adducts by immunogold electron microscopy showed appearance of 4HNE protein adducts in mitochondria as early as 3 hours, with a peak at 6 hours and subsequent decline at 24 hours. 3NT levels were significantly increased in all subcellular compartments at 6 hours and subsequently declined at 24 hours. Our data showed ADR induced 4HNE-protein adducts in mitochondria at the same time point as when mitochondrial injury initially appeared. These results document for the first time in vivo that mitochondrial oxidative damage precedes nitrative damage. The progressive nature of mitochondrial injury suggests that mitochondria, not other subcellular organelles, are the major site of intracellular injury.
Collapse
Affiliation(s)
- Luksana Chaiswing
- Department of Pathology and Laboratory Medicine, William S. Middleton Memorial Veterans Administration Hospital and University of Wisconsin Medical School, Madison WI 53705, USA
| | | | | | | | | | | |
Collapse
|
50
|
Meredith AM, Dass CR. Increasing role of the cancer chemotherapeutic doxorubicin in cellular metabolism. ACTA ACUST UNITED AC 2016; 68:729-41. [PMID: 26989862 DOI: 10.1111/jphp.12539] [Citation(s) in RCA: 201] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 02/05/2016] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The use of doxorubicin, a drug utilised for many years to treat a wide variety of cancers, has long been limited due to the significant toxicity that can occur not only during, but also years after treatment. It has multiple mechanisms of action including the intercalation of DNA, inhibition of topoisomerase II and the production of free radicals. We review the literature, with the aim of highlighting the role of drug concentration being an important determinant on the unfolding cell biological events that lead to cell stasis or death. METHODS The PubMed database was consulted to compile this review. KEY FINDINGS It has been found that the various mechanisms of action at the disposal of doxorubicin culminate in either cell death or cell growth arrest through various cell biological events, such as apoptosis, autophagy, senescence and necrosis. Which of these events is the eventual cause of cell death or growth arrest appears to vary depending on factors such as the patient, cell and cancer type, doxorubicin concentration and the duration of treatment. CONCLUSIONS Further understanding of doxorubicin's influence on cell biological events could lead to an improvement in the drug's efficacy and reduce toxicity.
Collapse
Affiliation(s)
| | - Crispin R Dass
- School of Pharmacy, Curtin University, Bentley, WA, Australia.,Curtin Biosciences Research Precinct, Bentley, WA, Australia
| |
Collapse
|