1
|
Gupta J, Sharma G. Nanogel: A versatile drug delivery system for the treatment of various diseases and their future perspective. Drug Deliv Transl Res 2025; 15:455-482. [PMID: 39103593 DOI: 10.1007/s13346-024-01684-w] [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] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
Nanogel (NG) drug delivery systems have emerged as promising tools for targeted and controlled drug release, revolutionizing treatment approaches across various diseases. Their unique physicochemical properties, such as nano size, high surface area, biocompatibility, stability, and tunable drug release, make them ideal carriers for a wide range of therapeutic agents. Nanogels (NGs), characterized by their 3D network of crosslinked polymers, offer unique edges like high drug loading capacity, controlled release, and targeted delivery. Additionally, the diverse applications of NGs in medical therapeutics highlight their versatility and potential impact on improving patient outcomes. Their application spans cancer treatment, infectious diseases, and chronic conditions, allowing for precise drug delivery to specific tissues or cells, minimizing side effects, and enhancing therapeutic efficacy. Despite their potential, challenges such as scalability, manufacturing reproducibility, and regulatory hurdles must be addressed. Achieving clinical translation requires overcoming these obstacles to ensure therapeutic payloads' safe and efficient delivery. Strategies such as surface modification and incorporating stimuli-responsive elements enhanced NG performance and addressed specific therapeutic challenges. Advances in nanotechnology, biomaterials, and targeted drug design offer opportunities to improve the performance of NGs and address current limitations. Tailoring NGs for exploring combination therapies and integrating diagnostics for real-time monitoring represent promising avenues for future research. In conclusion, NG drug delivery systems have demonstrated tremendous potential in diverse disease applications. Overcoming challenges and leveraging emerging technologies will pave the way for their widespread clinical implementation, ushering in a new era of precision medicine and improved patient care.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India.
| | - Gaurang Sharma
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India
| |
Collapse
|
2
|
Nguyen BL, Baumfalk DR, Lapierre-Nguyen SS, Zhong R, Doerr V, Montalvo RN, Wei-LaPierre L, Smuder AJ. Effects of exercise and doxorubicin on acute diaphragm neuromuscular transmission failure. Exp Neurol 2024; 378:114818. [PMID: 38782352 PMCID: PMC11616575 DOI: 10.1016/j.expneurol.2024.114818] [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: 02/06/2024] [Revised: 05/09/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
Doxorubicin (DOX) is a highly effective anthracycline antibiotic used to treat a wide variety of cancers including breast cancer, leukemia and lymphoma. Unfortunately, clinical use of DOX is limited due to adverse off-target effects resulting in fatigue, respiratory muscle weakness and dyspnea. The diaphragm is the primary muscle of inspiration and respiratory insufficiency is likely the result of both muscle weakness and neural impairment. However, the contribution of neuropathology to DOX-induced respiratory muscle dysfunction is unclear. We hypothesized that diaphragm weakness following acute DOX exposure is associated with neurotoxicity and that exercise preconditioning is sufficient to improve diaphragm muscle contractility by maintaining neuromuscular integrity. Adult female Sprague-Dawley rats were randomized into four experimental groups: 1) sedentary-saline, 2) sedentary-DOX, 3) exercise-saline or 4) exercise-DOX. Endurance exercise preconditioning consisted of treadmill running for 1 h/day at 30 m/min for 10 days. Twenty-four hours after the last bout of exercise, animals were treated with DOX (20 mg/kg, I.P.) or saline (equal volume). Our results demonstrate that 48-h following DOX administration diaphragm muscle specific force is reduced in sedentary-DOX rats in response to both phrenic nerve and direct diaphragm stimulation. Importantly, endurance exercise preconditioning in DOX-treated rats attenuated the decrease in diaphragm contractile function, reduced neuromuscular transmission failure and altered phrenic nerve morphology. These changes were associated with an exercise-induced reduction in circulating biomarkers of inflammation, nerve injury and reformation. Therefore, the results are consistent with exercise preconditioning as an effective way of reducing respiratory impairment via preservation of phrenic-diaphragm neuromuscular conduction.
Collapse
Affiliation(s)
- Branden L Nguyen
- Department Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd., Gainesville, FL 32611, United States of America.
| | - Dryden R Baumfalk
- Department Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd., Gainesville, FL 32611, United States of America
| | - Stephanie S Lapierre-Nguyen
- Department Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd., Gainesville, FL 32611, United States of America
| | - Renjia Zhong
- Department Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd., Gainesville, FL 32611, United States of America
| | - Vivian Doerr
- Department Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd., Gainesville, FL 32611, United States of America
| | - Ryan N Montalvo
- Department Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd., Gainesville, FL 32611, United States of America
| | - Lan Wei-LaPierre
- Department Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd., Gainesville, FL 32611, United States of America
| | - Ashley J Smuder
- Department Applied Physiology and Kinesiology, University of Florida, 1864 Stadium Rd., Gainesville, FL 32611, United States of America
| |
Collapse
|
3
|
Belhadjali F, Ghrir S, Ksia F, Limam F, Aouani E, Mokni M. Protective effect of grape seed extract and exercise training on tissues toxicities in doxorubicin-treated healthy rat. Biomarkers 2023; 28:544-554. [PMID: 37555371 DOI: 10.1080/1354750x.2023.2246698] [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: 06/13/2023] [Accepted: 08/06/2023] [Indexed: 08/10/2023]
Abstract
OBJECTIVE The aim of the present study was to investigate the effects of Grape seed extract (GSE) and exercise training on Doxorubicin (Doxo)-induced cardio, hepato and myo toxicities in healthy rats. METHODS Thirty male Wistar rats were randomly divided into five groups and daily treated by intraperitoneal route during two months either with ethanol 10% (Control); Doxo (1.5 mg/kg); Doxo + exercise (1.5 mg/kg + swimming exercise for 30 min twice a week); Doxo + GSE (1.5 mg/kg + GSE 2.5 g/kg); Doxo + GSE + exercise (1.5 mg/kg + GSE 2.5 g/kg + swimming exercise for 30 min twice a week). At the end of the treatment, tissues were collected and processed for the determination of oxidative stress (OS), intracellular mediators, energy fuelling biomarkers, carbohydrate metabolism parameters and muscle histopathology. RESULTS Doxo provoked OS characterised by an increased lipoperoxidation (LPO) and protein carbonylation and decreased antioxidant enzyme activities. Doxo also affected intracellular mediators, disturbed carbohydrate metabolism and energy fuelling in skeletal muscle as assessed by down-regulated Electron Transport Chain (ETC) complex activities leading in fine to altered skeletal muscle structure and function. CONCLUSION Almost all Doxo-induced disturbances were partially corrected with GSE and exercise on their own and more efficiently with the combined treatment (GSE + exercise).
Collapse
Affiliation(s)
- Feiza Belhadjali
- Laboratoire des Substances Bioactives, Centre de Biotechnologie, Technopole Borj-Cedria, Hammam-Lif, Tunisie
- Faculté des Sciences de Bizerte, Université de Carthage, Sidi Bou Saïd, Carthage, Tunisie
| | - Slim Ghrir
- Laboratoire des Substances Bioactives, Centre de Biotechnologie, Technopole Borj-Cedria, Hammam-Lif, Tunisie
| | - Féryel Ksia
- Laboratoire Environnement, Inflammation, Signalisation et Pathologies (LR 18ES40), Faculté de Médecine de Monastir, Université de Monastir, Monastir, Tunisie
| | - Ferid Limam
- Laboratoire des Substances Bioactives, Centre de Biotechnologie, Technopole Borj-Cedria, Hammam-Lif, Tunisie
| | - Ezzedine Aouani
- Laboratoire des Substances Bioactives, Centre de Biotechnologie, Technopole Borj-Cedria, Hammam-Lif, Tunisie
| | - Meherzia Mokni
- Laboratoire des Substances Bioactives, Centre de Biotechnologie, Technopole Borj-Cedria, Hammam-Lif, Tunisie
| |
Collapse
|
4
|
Li W, Cao J, Wang X, Zhang Y, Sun Q, Jiang Y, Yao J, Li C, Wang Y, Wang W. Ferruginol Restores SIRT1-PGC-1α-Mediated Mitochondrial Biogenesis and Fatty Acid Oxidation for the Treatment of DOX-Induced Cardiotoxicity. Front Pharmacol 2021; 12:773834. [PMID: 34899332 PMCID: PMC8652228 DOI: 10.3389/fphar.2021.773834] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Doxorubicin (DOX), a broad-spectrum chemotherapy drug, has life-threatening cardiotoxicity. Therefore, searching cardioprotective drugs for DOX-induced cardiotoxicity (DIC) is urgently needed. Objectives: This study aimed to explore cardioprotective effect and specific mechanism by which Ferruginol (FGL) attenuated DIC in vivo and in vitro. Methods: We evaluated the cardioprotection of FGL and performed high-throughput RNA-Seq on a DIC mouse. Whereafter, multiple methods, including western blot, RT-qPCR, a transmission electron microscope, CO-IP, immunofluorescence, and other staining methods, and antagonist of SIRT1 and PGC-1α were utilized to confirm the cardioprotection and molecular mechanism of FGL. Results: FGL-exerted cardioprotection manifested as enhanced cardiac function and reduced structural damage and apoptosis. The transcriptome and other results revealed that FGL facilitated PGC-1α-mediated mitochondrial biogenesis and fatty acid oxidation (MB and FAO) by increasing the expression of PGC-1α and concurrently promoting the expression of SIRT1-enhancing deacetylase SIRT1 deacetylating and activating PGC-1α. Conclusions: These results documented that FGL exerted cardioprotective effects restoring MB&FAO via the SIRT1–PGC-1α axis.
Collapse
Affiliation(s)
- Weili Li
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Cao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoping Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yawen Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qianbin Sun
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Yanyan Jiang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Junkai Yao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Beijing Key Laboratory of TCM Syndrome and Formula, Beijing, China.,Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, China
| |
Collapse
|
5
|
Mohebbati R, Jalili-Nik M, Saghi H, Sadatfaraji H, Soukhtanloo M. Zataria multiflora and its main ingredient, carvacrol, affect on the renal function, histopathological, biochemical and antioxidant parameters in adriamycin-induced nephrotic rats. Arch Physiol Biochem 2021; 127:453-461. [PMID: 31397187 DOI: 10.1080/13813455.2019.1650069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 06/27/2019] [Accepted: 07/25/2019] [Indexed: 10/26/2022]
Abstract
Oxidative stress has a major role in the nephrosis. In the present study, the effects of hydroalcoholic extract of Zataria multiflora (ZM) and carvacrol (CAR) were evaluated on the renal damage induced by adriamycin (ADR). The animals accidentally divided into four groups including: Control, ADR, ZM + ADR and CAR + ADR. The renal tissue, urine, and blood samples subjected to biochemical markers and histopathological evaluation. ADR significantly decreased glomerular filtration rate (GFR) while escalated urine protein excretion as well as protein clearance (p < .01 to p < .001). Also, ADR significantly reduced the antioxidants and boosted the malondialdehyde (MDA) compared to the control (p < .05 to p < .01). In groups treated by ZM and CAR, GFR, and antioxidants significantly increased, whereas urine protein excretion and MDA decreased (p < .05 to p < .001). ZM and CAR induced an improvement in ADR-induced renal damage by improving renal function as well as antioxidant activity.
Collapse
Affiliation(s)
- Reza Mohebbati
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Jalili-Nik
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Saghi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Sadatfaraji
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Soukhtanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Park J, Ahn S, Lee Y, Koh D, Lim Y. 1 H and 13 C NMR spectral assignment of 29 N'-(3-([1,1'-biphenyl]-4-yl)-1-phenyl-1H-pyrazol-4-yl)acylhydrazones. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2021; 59:648-662. [PMID: 33140870 DOI: 10.1002/mrc.5113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 06/11/2023]
Affiliation(s)
- Jihyun Park
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, South Korea
| | - Seunghyun Ahn
- Department of Applied Chemistry, Dongduk Women's University, Seoul, South Korea
| | - Youngshim Lee
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, South Korea
| | - Dongsoo Koh
- Department of Applied Chemistry, Dongduk Women's University, Seoul, South Korea
| | - Yoongho Lim
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul, South Korea
| |
Collapse
|
7
|
Sharma V, Gupta M, Kumar P, Sharma A. A Comprehensive Review on Fused Heterocyclic as DNA Intercalators: Promising Anticancer Agents. Curr Pharm Des 2021; 27:15-42. [PMID: 33213325 DOI: 10.2174/1381612826666201118113311] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/02/2020] [Indexed: 12/09/2022]
Abstract
Since the discovery of DNA intercalating agents (by Lerman, 1961), a growing number of organic, inorganic, and metallic compounds have been developed to treat life-threatening microbial infections and cancers. Fused-heterocycles are amongst the most important group of compounds that have the ability to interact with DNA. DNA intercalators possess a planar aromatic ring structure that inserts itself between the base pairs of nucleic acids. Once inserted, the aromatic structure makes van der Waals interactions and hydrogen-bonding interactions with the base pairs. The DNA intercalator may also contain an ionizable group that can form ionic interactions with the negatively charged phosphate backbone. After the intercalation, other cellular processes could take place, leading ultimately to cell death. The heterocyclic nucleus present in the DNA intercalators can be considered as a pharmacophore that plays an instrumental role in dictating the affinity and selectivity exhibited by these compounds. In this work, we have carried out a revision of small organic molecules that bind to the DNA molecule via intercalation and cleaving and exert their antitumor activity. A general overview of the most recent results in this area, paying particular attention to compounds that are currently under clinical trials, is provided. Advancement in spectroscopic techniques studying DNA interaction can be examined in-depth, yielding important information on structure-activity relationships. In this comprehensive review, we have focused on the introduction to fused heterocyclic agents with DNA interacting features, from medicinal point of view. The structure-activity relationships points, cytotoxicity data, and binding data and future perspectives of medicinal compounds have been discussed in detail.
Collapse
Affiliation(s)
- Vikas Sharma
- IIMT College of Pharmacy, Knowledge Park III, Greater Noida, Uttar Pradesh-201308, India
| | - Mohit Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Robertson Life Sciences Building, 2730 South Moody Avenue, Portland, OR 97201, United States
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 2193, South Africa
| | - Atul Sharma
- School of Chemistry, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
8
|
Pharmacokinetics of the intraperitoneal nanoparticle pegylated liposomal doxorubicin in patients with peritoneal metastases. Eur J Surg Oncol 2021; 47:108-114. [DOI: 10.1016/j.ejso.2019.03.035] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/11/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
|
9
|
Huertas AM, Morton AB, Hinkey JM, Ichinoseki-Sekine N, Smuder AJ. Modification of Neuromuscular Junction Protein Expression by Exercise and Doxorubicin. Med Sci Sports Exerc 2020; 52:1477-1484. [PMID: 31985575 DOI: 10.1249/mss.0000000000002286] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Doxorubicin (DOX) is a highly effective antitumor agent widely used in cancer treatment. However, it is well established that DOX induces muscular atrophy and impairs force production. Although no therapeutic interventions exist to combat DOX-induced muscle weakness, endurance exercise training has been shown to reduce skeletal muscle damage caused by DOX administration. Numerous studies have attempted to identify molecular mechanisms responsible for exercise-induced protection against DOX myotoxicity. Nevertheless, the mechanisms by which endurance exercise protects against DOX-induced muscle weakness remain elusive. In this regard, impairments to the neuromuscular junction (NMJ) are associated with muscle wasting, and studies indicate that physical exercise can rescue NMJ fragmentation. Therefore, we tested the hypothesis that exercise protects against DOX-induced myopathy by preventing detrimental changes to key proteins responsible for maintenance of the NMJ. METHODS Female Sprague-Dawley rats were assigned to sedentary or exercise-trained groups. Exercise training consisted of a 5-d treadmill habituation period followed by 10 d of running (60 min·d, 30 m·min, 0% grade). After the last training bout, exercise-trained and sedentary animals were paired with either placebo (saline) or DOX (20 mg·kg i.p.) treatment. Two days after drug treatment, the soleus muscle was excised for subsequent analyses. RESULTS Our results indicate that endurance exercise training prevents soleus muscle atrophy and contractile dysfunction in DOX-treated animals. These adaptations were associated with the increased expression of the following neurotrophic factors: brain-derived neurotrophic factor, glial cell line-derived neurotrophic factor, nerve growth factor, and neurotrophin-3. In addition, exercise enhanced the expression of receptor-associated protein of the synapse and the acetylcholine receptor (AChR) subunits AChRβ, AChRδ, and AChRγ in DOX-treated animals. CONCLUSION Therefore, upregulating neurotrophic factor and NMJ protein expression may be an effective strategy to prevent DOX-induced skeletal muscle dysfunction.
Collapse
Affiliation(s)
- Andres Mor Huertas
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | - Aaron B Morton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| | | | | | - Ashley J Smuder
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL
| |
Collapse
|
10
|
Noll NA, Lal H, Merryman WD. Mouse Models of Heart Failure with Preserved or Reduced Ejection Fraction. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1596-1608. [PMID: 32343958 DOI: 10.1016/j.ajpath.2020.04.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022]
Abstract
Heart failure (HF) is a chronic, complex condition with increasing incidence worldwide, necessitating the development of novel therapeutic strategies. This has led to the current clinical strategies, which only treat symptoms of HF without addressing the underlying causes. Multiple animal models have been developed in an attempt to recreate the chronic HF phenotype that arises following a variety of myocardial injuries. Although significant strides have been made in HF research, an understanding of more specific mechanisms will require distinguishing models that resemble HF with preserved ejection fraction (HFpEF) from those with reduced ejection fraction (HFrEF). Therefore, current mouse models of HF need to be re-assessed to determine which of them most closely recapitulate the specific etiology of HF being studied. This will allow for the development of therapies targeted specifically at HFpEF or HFrEF. This review will summarize the commonly used mouse models of HF and discuss which aspect of human HF each model replicates, focusing on whether HFpEF or HFrEF is induced, to allow better investigation into pathophysiological mechanisms and treatment strategies.
Collapse
Affiliation(s)
- Natalie A Noll
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Hind Lal
- Department of Medicine, Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee.
| |
Collapse
|
11
|
Lambertenghi-Deliliers G, Zanon PL, Pozzoli EF, Bellini O. Myocardial Injury Induced by a Single Dose of Adriamycin: An Electron Microscopic Study. TUMORI JOURNAL 2018; 62:517-28. [PMID: 1020054 DOI: 10.1177/030089167606200506] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adriamycin cardiomyopathy has been studied under the electron microscope using myocardial ventricular cells of CRF mice, previously treated with 10 mg/kg body weight of the drug given in a single intravenous injection. Within 10 min myocardial cell nucleoli show a nucleolonema fragmentation, and during the following 3 hours they acquire the nucleolar segregation pattern. Fourteen hours after drug injection, nucleolar morphology again becomes normal, while areas of focal degeneration, characterized by damaged mitochondria and enlarged smooth reticulum cisternae, appear in the sarcoplasm. One to 3 days later the degeneration process involves the myofibrillar component, and after 50 days the great majority of myocardial ventricular cells is damaged. The early appearance and the functional significance of nucleolar segregation support the hypothesis that adriamycin cardiotoxicity might be dependent on its ability to bind to myocardial cell DNA. The consequent failure of UNA and protein synthesis, impairing the continuous renewal of myofibrillar and mitochondrial components of the cell, might explain the progressive myocardial damage.
Collapse
|
12
|
Chitnis M, Adwankar M. Potentiation of Adriamycin Cytotoxicity in P388 Murine Leukemia Sensitive and Resistant to Adriamycin by use of Lonidamine and Hyperthermia. TUMORI JOURNAL 2018; 72:469-73. [PMID: 3798566 DOI: 10.1177/030089168607200503] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The in vitro effect of adriamycin (ADR) and lonidamine alone and in combination, at 37 °C and 43 °C, was investigated on murine leukemia P388 sensitive (P388/S) and resistant (P388/ADR) to adriamycin. The sensitive and the resistant cells were exposed in vitro with and without the drugs for 1 h at 37 °C and 43 °C. These cells were inoculated ip (106 cells/mouse) into groups of BDF, mice. Cytotoxic effect of the treatment was assessed on the basis of percentage increase in life span (% ILS) of these animals, compared to the animals receiving cells which did not receive any treatment but exposed only to 37 °C for 1 h. It was observed that exposure of P388/ADR cells to lonidamine or adriamycin alone at 43 °C for 1 h resulted in greater cell kill, thus enhancing the % ILS of the experimental animals receiving those cells, compared to that of mice receiving the cells exposed to the same drugs for 1 h at 37 °C. However, the combination of lonidamine (0.02 mM) and adriamycin (10 μg/ml) at 43 °C for 1 h showed more than a synergistic effect, resulting in a % ILS of 120. Similar results were seen in the case of P388/S; however, the observations pertaining to P388/ADR are encouraging, since the mode of treatment has reversed the acquired resistance of P388 leukemia cells to adriamycin.
Collapse
|
13
|
Chitnis MP, Menon RS, Gude RP. Effect of Tween 80 on Adriamycin Cytotoxicity in Murine P388 Leukemia. TUMORI JOURNAL 2018; 70:313-5. [PMID: 6474580 DOI: 10.1177/030089168407000403] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Studies on mice bearing P388 adriamycin sensitive and resistant lymphocytic leukemia were carried out to compare antitumor activity of adriamycin dissolved in Tween 80 (10 % in distilled water) with that of adriamycin dissolved in distilled water alone. The antitumor activity of adriamycin dissolved in acqueous solutions of Tween 80 was higher than that of adriamycin dissolved in distilled water against P388 leukemia sensitive to adriamycin. However, no change in the activity was observed in P388 leukemia resistant to adriamycin when the drug was administered either in acqueous solution of Tween 80 or in distilled water alone.
Collapse
|
14
|
Parekh H, Chavan S, Chitnis M. Antiproliferative Effects of Mitoxantrone in Adr-Sensitive and Adr-Resistant P388 Leukemia Cells Enhanced by Vitamin K3. TUMORI JOURNAL 2018; 77:484-90. [PMID: 1803714 DOI: 10.1177/030089169107700607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Vitamin K3 was employed as a resistance-modifying agent to Investigate its activity in enhancing mitoxantrone (MITO)-induced cytotoxicity in parental (P388/S) and multidrug resistant (P388/ADR) P388 leukemia cells. Vitamin K3 potentiated the antitumor effects of MITO in P388/S and P388/ ADR tumor cells as monitored by inhibition of tumor cell survival (MTT assay). MITO and vitamin K3 in combination effected an enhanced inhibition of [3H]thymidine (DNA synthesis) and [3H]uridine (RNA synthesis) and also Increased the life span of the sensitive and resistant tumor-bearing animals. The effect of vitamin K3 on the induction of DNA strand breaks by MITO was also examined. Increased fragmentation of DNA was illustrated in the sensitive and resistant P388 leukemia cells exposed to the combination. Observations indicate the restoration of sensitivity in P388/ADR cells to MITO by vitamin K3 that may be due to its ability to increase the MITO-induced DNA strand breaks.
Collapse
Affiliation(s)
- H Parekh
- Chemotherapy Division, Cancer Research Institute, Tata Memorial Centre, Parel, Bombay, India
| | | | | |
Collapse
|
15
|
Kolarić K, Maricić Z, Roth A, Dujmović I. Adriamycin Alone and in Combination with Radiotherapy in the Treatment of Inoperable Esophageal Cancer. TUMORI JOURNAL 2018; 63:485-91. [PMID: 601878 DOI: 10.1177/030089167706300510] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Thirty-three patients with inoperable esophageal cancer were treated in a randomized study. There were 24 squamous cell, 2 anaplastic, 4 unclassified, and 3 adenocarcinomas. Eighteen patients were treated with 6 cycles of adriamycin with a 3 week rest period between cycles. Adriamycin was administered in doses of 40 mg/m2 body surface daily for 2 days during each cycle. The other group of 15 patients were treated with both adriamycin and radiation. The tumor was irradiated with speed electrons (4500–5200 rads total dose), and during irradiation, 3 cycles of adriamycin were given. All patients previously had had no treatment. In the group of patients treated with adriamycin, the response rate (> 50 % tumor regression) was 33 % (6/18) with 1 complete and 5 partial remissions. In the combined treatment group, there were 4 complete and 5 partial remissions (9/15) with a response rate of 60 %. In both groups of patients, remissions were obtained in the squamous cell carcinomas, except 1 patient with anaplastic carcinoma in the adriamycin-treated group, and 1 adenocarcinoma in the combined treated group. Remissions lasted 2–12 months (M = 3.2 months) in the adriamycin-treated group and 3–15 months (M = 8.6 months) in the combined treatment group. There were no significant differences in the toxic side effects, except a slightly increased myelodepression in the combined treatment modality. One patient of the combined treatment group showed a potentiation of radiation dermititis. The results obtained show that adriamycin does have an antitumor effect on esophageal (squamous cell) carcinoma, which so far has not been proven. Significantly better results with longer remission duration can be obtained when both adriamycin and radiation are combined.
Collapse
|
16
|
Casazza AM, Pratesi G, Giuliani F, Formelli F, Di Marco A. Enhancement of the Antitumor Activity of Adriamycin by Tween 80. TUMORI JOURNAL 2018; 64:115-29. [PMID: 675847 DOI: 10.1177/030089167806400202] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
This paper describes the effect of Tween 80 on the antitumor activity and on the distribution of adriamycin in mice. The dilution of adriamycin in a 10% water solution of Tween 80 produced a significant increase of the antitumor activity in mice against ascites tumors (L1210 leukemia), disseminated leukemias (transplanted leukemias originally induced by Gross leukemia virus and Moloney leukemia virus), and solid tumors (Sarcoma 180, MS-2 sarcoma). In all these experiments the drug was administered i.v., according to different schedules. Higher antitumor activity at the optimal dose and an increase of activity at lower doses were observed in different experimental systems. Toxicity was also slightly enhanced. Tissue distribution was studied in normal mice and in tumor-bearing mice (Gross leukemia and MS-2 sarcoma). In animals given i.v. adriamycin diluted in 10% Tween 80 there was a higher drug concentration in spleen, lung and kidney than there was in mice given the drug in a water solution. In all the other organs examined (heart, liver, small intestine) and in the MS-2 tumor tissue, no significant increase was observed. In L1210 leukemia-bearing mice, i.p. treatment with adriamycin diluted in 10% Tween 80 resulted in a significantly higher toxicity than that which resulted from treatment with adriamycin in a water solution; no increase of antitumor activity was observed.
Collapse
|
17
|
Lambertenghi-Deliliers G, Zanon PL, Pozzoli EF, Bellini O, Praga C. Ultrastructural Alterations of Atrial Myocardium Induced by Adriamycin in Chronically Treated Animals. TUMORI JOURNAL 2018; 64:15-24. [PMID: 565963 DOI: 10.1177/030089167806400102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The clinical use of adriamycin (AM) is limited by a possible dose-dependent myocardiopathy. Severe lesions of ventricular myocardium widely described by electron microscopy have been correlated to irreversible congestive heart failure. On the other hand, the atrial contractile elements which differ from the ventricular ones because of the presence of the so-called specific granules have rarely been considered. In the work described in this paper, adriamycin was injected into rabbits and mice according to schedules of chronic toxicity. At the end of the treatment the atrial myocells presented diffuse ultrastructural lesions of mitochondria, sarcoplasmic reticulum and myofibrillar bundles. These alterations might be caused by the ribonucleoprotein synthesis inhibition, by a direct drug toxicity or by an energetic crisis due to early mitochondrial lesions. Besides, adriamycin produces a decrease of the specific atrial granules that play a hypothetic role in the metabolism of myocardial cells. However, lack of information about the contents and the exact function of atrial granules does not allow us to conclude that their decrease in treated animals has a pathogenetic significance in myocardiopathy induced by adriamycin.
Collapse
|
18
|
Congras A, Caillet N, Torossian N, Quelen C, Daugrois C, Brousset P, Lamant L, Meggetto F, Hoareau-Aveilla C. Doxorubicin-induced loss of DNA topoisomerase II and DNMT1- dependent suppression of MiR-125b induces chemoresistance in ALK-positive cells. Oncotarget 2018; 9:14539-14551. [PMID: 29581862 PMCID: PMC5865688 DOI: 10.18632/oncotarget.24465] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 11/20/2017] [Indexed: 12/04/2022] Open
Abstract
Systemic anaplastic large-cell lymphoma (ALCL) is a childhood T cell neoplasm defined by the presence or absence of translocations that lead to the ectopic expression of anaplastic lymphoma kinase (ALK), with nucleophosmin-ALK (NPM-ALK) fusions being the most common. Polychemotherapy involving doxorubicin is the standard first-line treatment but for the 25 to 35% of patients who relapse and develop resistance the prognosis remains poor. We studied the potential role of the microRNA miR-125b in the development of resistance to doxorubicin in NPM-ALK(+) ALCL. Our results show that miR-125b expression is repressed in NPM-ALK(+) cell lines and patient samples through hypermethylation of its promoter. NPM-ALK activity, in cooperation with DNA topoisomerase II (Topo II) and DNA methyltransferase 1 (DNMT1), is responsible for miR-125b repression through DNA hypermethylation. MiR-125b repression was reversed by the inhibition of DNMTs with decitabine or the inhibition of DNA topoisomerase II with either doxorubicin or etoposide. In NPM-ALK(+) cell lines, doxorubicin treatment led to an increase in miR-125b levels by inhibiting the binding of DNMT1 to the MIR125B1 promoter and downregulating the pro-apoptotic miR-125b target BAK1. Reversal of miR-125b silencing, increased miR-125b levels and reduced BAK1 expression also led to a lower efficacy of doxorubicin, suggestive of a pharmacoresistance mechanism. In line with this, miR-125b repression and increased BAK1 expression correlated with early relapse in human NPM-ALK(+) ALCL primary biopsies. Collectively our findings suggest that miR-125b could be used to predict therapeutic outcome in NPM-ALK(+) ALCL.
Collapse
Affiliation(s)
- Annabelle Congras
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 CRCT, F-31000 Toulouse, France.,Equipe Labelisée LIGUE 2017
| | - Nina Caillet
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 CRCT, F-31000 Toulouse, France.,Equipe Labelisée LIGUE 2017
| | - Nouritza Torossian
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 CRCT, F-31000 Toulouse, France
| | - Cathy Quelen
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 CRCT, F-31000 Toulouse, France.,Equipe Labelisée LIGUE 2017
| | - Camille Daugrois
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 CRCT, F-31000 Toulouse, France
| | - Pierre Brousset
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 CRCT, F-31000 Toulouse, France.,Institut Carnot Lymphome-CALYM, 31024, Toulouse, France.,Laboratoire d'Excellence Toulouse Cancer-TOUCAN, 31024, Toulouse, France.,European Research Initiative on ALK-related malignancies (ERIA) (http://www.erialcl.net/).,Equipe Labelisée LIGUE 2017
| | - Laurence Lamant
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 CRCT, F-31000 Toulouse, France.,Institut Carnot Lymphome-CALYM, 31024, Toulouse, France.,Laboratoire d'Excellence Toulouse Cancer-TOUCAN, 31024, Toulouse, France.,European Research Initiative on ALK-related malignancies (ERIA) (http://www.erialcl.net/).,Equipe Labelisée LIGUE 2017
| | - Fabienne Meggetto
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 CRCT, F-31000 Toulouse, France.,Institut Carnot Lymphome-CALYM, 31024, Toulouse, France.,Laboratoire d'Excellence Toulouse Cancer-TOUCAN, 31024, Toulouse, France.,European Research Initiative on ALK-related malignancies (ERIA) (http://www.erialcl.net/).,Equipe Labelisée LIGUE 2017
| | - Coralie Hoareau-Aveilla
- Inserm, UMR1037 CRCT, F-31000 Toulouse, France.,Université Toulouse III-Paul Sabatier, UMR1037 CRCT, F-31000 Toulouse, France.,CNRS, ERL5294 CRCT, F-31000 Toulouse, France.,Equipe Labelisée LIGUE 2017
| |
Collapse
|
19
|
Liu Z, Liu J, Wang T, Li Q, Francis PS, Barrow CJ, Duan W, Yang W. Switching off the interactions between graphene oxide and doxorubicin using vitamin C: combining simplicity and efficiency in drug delivery. J Mater Chem B 2018; 6:1251-1259. [DOI: 10.1039/c7tb03063k] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Delivery of doxorubicin using graphene oxide is remarkably improved by adding a little amount of vitamin C.
Collapse
Affiliation(s)
- Zhen Liu
- Centre for Chemistry and Biotechnology
- School of Life and Environmental Sciences
- Deakin University
- Geelong
- Australia
| | - Jingquan Liu
- College of Chemical Science and Engineering
- Laboratory of Fiber Materials and Modern Textile
- The Growing Base for State Key Laboratory
- Qingdao University
- Qingdao
| | - Tao Wang
- School of Nursing
- Zhengzhou University
- Zhengzhou
- China
- School of Medicine
| | - Qiong Li
- School of Medicine
- Faculty of Health
- Deakin University
- Geelong
- Australia
| | - Paul S. Francis
- Centre for Chemistry and Biotechnology
- School of Life and Environmental Sciences
- Deakin University
- Geelong
- Australia
| | - Colin J. Barrow
- Centre for Chemistry and Biotechnology
- School of Life and Environmental Sciences
- Deakin University
- Geelong
- Australia
| | - Wei Duan
- School of Medicine
- Faculty of Health
- Deakin University
- Geelong
- Australia
| | - Wenrong Yang
- Centre for Chemistry and Biotechnology
- School of Life and Environmental Sciences
- Deakin University
- Geelong
- Australia
| |
Collapse
|
20
|
Comparision of doxorubicin-induced cardiotoxicity in the ICR mice of different sources. Lab Anim Res 2017; 33:165-170. [PMID: 28747983 PMCID: PMC5527143 DOI: 10.5625/lar.2017.33.2.165] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 01/05/2023] Open
Abstract
Doxorubicin is a widely used chemotherapeutic agents and is now part of standard therapeutic regimens for a variety of cancers (eg, hematopoietic malignancies and advanced solid tumors of the breast, ovary, thyroid, and bone). However, a potentially lethal and dose-dependent cardiotoxicity that appears within a short time after treatment limits the usage of doxorubicin in cancer patients. Although the mechanism of doxorubicin-induced cardiotoxicity is not completely understood, it is thought that free radical-induced oxidative stress and excessive production of reactive oxygen species are primary drivers of its toxicity. In this study, we compared the doxorubicin-induced cardiotoxicity of ICR mice obtained from three different sources and evaluated the utility of Korl:ICR stock established by the Korean FDA. Because doxorubicin-induced cardiotoxicity is thought to involve the excessive generation of ROS followed by oxidative stress, we determined the representative tissue index of oxidation, lipid peroxidation, and antioxidant, glutathione (GSH), as well as the parameters of heart injury. Doxorubicin treatment successfully induced cardiotoxicity as evidenced by histological examination and serum parameters (eg, levels of LDH and CK activities) in ICR mice. It was accompanied by increased lipid peroxidation and a decrease in both cysteine and GSH, further supporting previous reports that oxidative stress is a potential mechanism of doxorubicin-induced cardiotoxicity. Of interest, we did not observe a significant difference in doxorubicin-induced cardiotoxicity among mice of different origins. Collectively, our results suggest that Korl:ICR strain may be useful in the research of doxorubicin-induced cardiotoxicity.
Collapse
|
21
|
Muramatsu Y, Matsui T, Deie M, Sato K. Pulsed Electromagnetic Field Stimulation Promotes Anti-cell Proliferative Activity in Doxorubicin-treated Mouse Osteosarcoma Cells. ACTA ACUST UNITED AC 2017; 31:61-68. [PMID: 28064222 DOI: 10.21873/invivo.11026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/14/2016] [Accepted: 11/23/2016] [Indexed: 01/21/2023]
Abstract
AIM We aimed to investigate the synergistic effects of pulsed electromagnetic field (PEMF) and doxorubicin therapy in a mouse osteosarcoma cell line (LM8 cells) in vitro. MATERIALS AND METHODS The effects of PEMF (5 mT, 200 Hz) of different durations and doxorubicin on the proliferative activity of LM8 cells were measured by the MTT assay. Apoptotic-related factors such as cell-cycle phase, mitochondrial membrane potential, and caspase 3/7 activity were investigated using 4',6-diamidino-2-phenylindole staining and apoptosis kits. Identification of intracellular signaling molecules induced by the combination was comprehensively explored using a stress and apoptosis-related protein array kit. RESULTS PEMF enhanced the inhibition of cell proliferation mediated by doxorubicin but did not affect the cell cycle, mitochondrial membrane potential, or doxorubicin-induced G2/M arrest. The combination of PEMF and doxorubicin altered a few signaling molecules. PEMF tended to reduce the doxorubicin-induced decrease of phosphorylated BAD, while reducing the increased expression of total IĸB and phosphorylated-CHK1 induced by doxorubicin. CONCLUSION Our results indicate that combination of PEMF and doxorubicin could be a novel chemotherapeutic strategy.
Collapse
Affiliation(s)
- Yoshitaka Muramatsu
- Department of Orthopaedic Surgery, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| | - Takuya Matsui
- Department of Physiology, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| | - Masataka Deie
- Department of Orthopaedic Surgery, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| | - Keiji Sato
- Department of Orthopaedic Surgery, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| |
Collapse
|
22
|
Sintov AC, Enden G. New doxorubicin nanoparticles engineered from calcium-crosslinked carbomer and a microemulsion precursor. Drug Dev Ind Pharm 2016; 43:830-838. [DOI: 10.1080/03639045.2016.1239730] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
23
|
Doxorubicin chemotherapy affects intracellular and interstitial nitric oxide concentrations in skeletal muscle. Cell Biol Toxicol 2016; 32:121-31. [DOI: 10.1007/s10565-016-9325-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 03/30/2016] [Indexed: 10/22/2022]
|
24
|
Eisa NH, ElSherbiny NM, Shebl AM, Eissa LA, El-Shishtawy MM. Phenethyl isothiocyanate potentiates anti-tumour effect of doxorubicin through Akt-dependent pathway. Cell Biochem Funct 2015; 33:541-551. [PMID: 26548747 DOI: 10.1002/cbf.3153] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 04/03/2025]
Abstract
The present study aims to investigate the in vivo and in vitro anti-tumour properties of phenethyl isothiocyanate (PEITC) alone and in combination with doxorubicin (Dox). The anti-tumour activity was evaluated in vitro by MTT assay using cultured human breast cancer cell line (MCF-7) and human hepatoma cell line (HepG-2) cell lines. In vivo, Ehrlich solid tumour model was used. Tumour volume, weight and antioxidant parameters were determined. Immunohistochemistry analysis for active (cleaved) caspase-3 was also performed. We tested the effect of PEITC treatment on pAkt/Akt ratio, NF-κB p65 DNA binding activity and caspase-9 enzyme activity in both MCF-7 and HepG-2 cell lines. Effect of PEITC treatment on cell migration was assessed by wound healing assay. PEITC and/or Dox treatment significantly inhibited solid tumour volume and tumour weight when compared with control mice. PEITC treatment significantly reduced oxidative stress caused by Dox treatment as indicated by significant increase in total antioxidant capacity and decrease in malondialdehyde level. Microscopic examination of tumour tissues showed a significant increase in active (cleaved) caspase-3 expression in PEITC and/or Dox treated groups. PEITC showed a dose-dependent inhibition of MCF-7 and HepG-2 cellular viability. PEITC inhibited Akt and NF-κB activation and increased caspase-9 activity in a dose-dependent manner. PEITC treatment effectively inhibited both MCF-7 and HepG-2 cell migration. We can conclude that PEITC acts via multiple molecular targets to elicit anti-carcinogenic activity. PEITC/Dox combination therapy might be a potential novel strategy, which may benefit patients with breast and liver cancers.
Collapse
Affiliation(s)
- Nada H Eisa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Nehal M ElSherbiny
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Abdelhadi M Shebl
- Department of Pathology, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Laila A Eissa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Mamdouh M El-Shishtawy
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
25
|
Sinha BK, Mason RP. IS METABOLIC ACTIVATION OF TOPOISOMERASE II POISONS IMPORTANT IN THE MECHANISM OF CYTOTOXICITY? ACTA ACUST UNITED AC 2015; 6. [PMID: 31171989 DOI: 10.4172/2157-7609.1000186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The antitumor drugs doxorubicin and etoposide, a phodophyllotoxin derivative, are clinically active for the treatment of human malignancies. Because of their extreme effectiveness in the clinic, their modes of actions have been the subject of intense research for over several decades both in the laboratory and in the clinic. It has been found that both doxorubicin and etoposide (VP-16) act on topoisomerase II, induce DNA cleavage, and form double-strand breaks, causing tumor cell death. However, both of these drugs also undergo extensive metabolism in tumor cells and in vivo to various reactive intermediates that bind covalently to cellular DNA and proteins. Moreover, both drugs are metabolized to reactive free radicals that induce lipid peroxidation and DNA damage. However, the role of drug activation in the mechanism of cytotoxicity remains poorly defined. In this review, we critically evaluate the significance of metabolic activation of doxorubicin and etoposide in the mechanism of tumor cytotoxicity.
Collapse
Affiliation(s)
- Birandra K Sinha
- Immunity, Inflammation and Disease Laboratory, National Institutes of Environmental Health Sciences, NIH, Research Triangle, Park, North Carolina, USA
| | - Ronald P Mason
- Immunity, Inflammation and Disease Laboratory, National Institutes of Environmental Health Sciences, NIH, Research Triangle, Park, North Carolina, USA
| |
Collapse
|
26
|
Caviglia C, Zór K, Canepa S, Carminati M, Larsen LB, Raiteri R, Andresen TL, Heiskanen A, Emnéus J. Interdependence of initial cell density, drug concentration and exposure time revealed by real-time impedance spectroscopic cytotoxicity assay. Analyst 2015; 140:3623-9. [PMID: 25868456 DOI: 10.1039/c5an00097a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We investigated the combined effect of the initial cell density (12,500, 35,000, 75,000, and 100,000 cells cm(-2)) and concentration of the anti-cancer drug doxorubicin on HeLa cells by performing time-dependent cytotoxicity assays using real-time electrochemical impedance spectroscopy. A correlation between the rate of cell death and the initial cell seeding density was found at 2.5 μM doxorubicin concentration, whereas this was not observed at 5 or 100 μM. By sensing the changes in the cell-substrate interaction using impedance spectroscopy under static conditions, the onset of cytotoxicity was observed 5 h earlier than when using a standard colorimetric end-point assay (MTS) which measures changes in the mitochondrial metabolism. Furthermore, with the MTS assay no cytotoxicity was observed after 15 h of incubation with 2.5 μM doxorubicin, whereas the impedance showed at this time point cell viability that was below 25%. These results indicate that impedance detection reveals cytotoxic events undetectable when using the MTS assay, highlighting the importance of combining impedance detection with traditional drug toxicity assays towards a more in depth understanding of the effect of anti-cancer drugs on in vitro assays. Moreover, the detection of doxorubicin induced toxicity determined with impedance under static conditions proved to be 6 times faster than in perfusion culture.
Collapse
Affiliation(s)
- C Caviglia
- Department of Micro- and Nanotechnology, Technical University of Denmark, Kgs. Lyngby, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Caviglia C, Zór K, Montini L, Tilli V, Canepa S, Melander F, Muhammad HB, Carminati M, Ferrari G, Raiteri R, Heiskanen A, Andresen TL, Emnéus J. Impedimetric toxicity assay in microfluidics using free and liposome-encapsulated anticancer drugs. Anal Chem 2015; 87:2204-12. [PMID: 25582124 DOI: 10.1021/ac503621d] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
In this work, we have developed a microfluidic cytotoxicity assay for a cell culture and detection platform, which enables both fluid handling and electrochemical/optical detection. The cytotoxic effect of anticancer drugs doxorubicin (DOX), oxaliplatin (OX) as well as OX-loaded liposomes, developed for targeted drug delivery, was evaluated using real-time impedance monitoring. The time-dependent effect of DOX on HeLa cells was monitored and found to have a delayed onset of cytotoxicity in microfluidics compared with static culture conditions based on data obtained in our previous study. The result of a fluorescent microscopic annexin V/propidium iodide assay, performed in microfluidics, confirmed the outcome of the real-time impedance assay. In addition, the response of HeLa cells to OX-induced cytotoxicity proved to be slower than toxicity induced by DOX. A difference in the time-dependent cytotoxic response of fibrosarcoma cells (HT1080) to free OX and OX-loaded liposomes was observed and attributed to incomplete degradation of the liposomes, which results in lower drug availability. The matrix metalloproteinase (MMP)-dependent release of OX from OX-loaded liposomes was also confirmed using laryngopharynx carcinoma cells (FaDu). The comparison and the observed differences between the cytotoxic effects under microfluidic and static conditions highlight the importance of comparative studies as basis for implementation of microfluidic cytotoxic assays.
Collapse
Affiliation(s)
- Claudia Caviglia
- Department of Micro- and Nanotechnology, Technical University of Denmark , DK-2800 Kongens Lyngby, Denmark
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ahmed SE, Martins AM, Husseini GA. The use of ultrasound to release chemotherapeutic drugs from micelles and liposomes. J Drug Target 2014; 23:16-42. [PMID: 25203857 DOI: 10.3109/1061186x.2014.954119] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Several drug delivery systems have been investigated to reduce the side effects of chemotherapy by encapsulating the therapeutic agent in a nanosized carrier until it reaches the tumor site. Many of these particles are designed to be responsive to the mechanical and thermal perturbations delivered by ultrasound. Once the nanoparticle reaches the desired location, ultrasound is applied to release the chemotherapy drug only in the vicinity of the targeted (cancer) site, thus avoiding any detrimental interaction with healthy cells in the body. Studies using liposomes and micelles have shown promising results in this area, as these nanoparticles with simple, yet effective structures, showed high efficiency as drug delivery vehicles both in vitro and in vivo. This article reviews the design and application of two novel nanosized chemotherapeutic carriers (i.e. micelles and liposomes) intended to be actuated by ultrasound.
Collapse
Affiliation(s)
- Salma E Ahmed
- Department of Chemical Engineering, American University of Sharjah , Sharjah , United Arab Emirates
| | | | | |
Collapse
|
29
|
Kim KB, Yang JY, Kwack SJ, Kim HS, Ryu DH, Kim YJ, Bae JY, Lim DS, Choi SM, Kwon MJ, Bang DY, Lim SK, Kim YW, Hwang GS, Lee BM. Potential metabolomic biomarkers for evaluation of adriamycin efficacy using a urinary 1H-NMR spectroscopy. J Appl Toxicol 2013; 33:1251-1259. [PMID: 22782856 DOI: 10.1002/jat.2778] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 04/19/2012] [Accepted: 04/19/2012] [Indexed: 12/16/2022]
Abstract
A metabolomics approach using proton nuclear magnetic resonance (NMR) was applied to investigate metabolic alterations following adriamycin (ADR) treatment for gastric adenocarcinoma. After BALB/c-nu/nu mice were implanted with human gastric adenocarcinoma, ADR (1 or 3 mg kg(-1) per day) was intraperitoneally administered for 5 days. Urine was collected on days 2 and 5 and analyzed by NMR. The levels of trimethylamine oxide (TMAO, ×0.3), hippurate (×0.3) and taurine (×0.6) decreased significantly (P < 0.05), whereas the levels of 3-indoxylsulfate (×12.6), trigonelline (×1.5), citrate (×2.5), trimethylamine (TMA, ×2.0) and 2-oxoglutarate (×2.3) increased significantly (P < 0.05) in the tumor model. After ADR treatment, TMAO, hippuarte and taurine were increased significantly on day 5 compared with those of the tumor model. The levels of 2-oxoglutarate, 3-indoxylsulfate, trigonelline, TMA and citrate, which increased in the tumor model, significantly decreased to those of normal control by ADR treatment. Furthermore, the ratio between TMA and TMAO was dramatically altered in both tumor and ADR-treated groups. Overall, metabolites such as TMAO, TMA, 3-indoxylsulfate, hippurate, trigonelline, citrate and 2-oxoglutarate related to the tricarboxylic acid (TCA) cycle might be considered as therapeutic targets to potentiate the efficacy of ADR. Thus, these results suggest that the metabolomics analysis of tumor response to ADR treatment may be applicable for demonstrating the efficacy of anticancer agent, ADR and treatment adaptation.
Collapse
Affiliation(s)
- Kyu-Bong Kim
- College of Pharmacy, Dankook University, Dandae-ro, Cheonan, Chungnam, 330-714, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Enhancing cell nucleus accumulation and DNA cleavage activity of anti-cancer drug via graphene quantum dots. Sci Rep 2013; 3:2852. [PMID: 24092333 PMCID: PMC3790198 DOI: 10.1038/srep02852] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/10/2013] [Indexed: 02/07/2023] Open
Abstract
Graphene quantum dots (GQDs) maintain the intrinsic layered structural motif of graphene but with smaller lateral size and abundant periphery carboxylic groups, and are more compatible with biological system, thus are promising nanomaterials for therapeutic applications. Here we show that GQDs have a superb ability in drug delivery and anti-cancer activity boost without any pre-modification due to their unique structural properties. They could efficiently deliver doxorubicin (DOX) to the nucleus through DOX/GQD conjugates, because the conjugates assume different cellular and nuclear internalization pathways comparing to free DOX. Also, the conjugates could enhance DNA cleavage activity of DOX markedly. This enhancement combining with efficient nuclear delivery improved cytotoxicity of DOX dramatically. Furthermore, the DOX/GQD conjugates could also increase the nuclear uptake and cytotoxicity of DOX to drug-resistant cancer cells indicating that the conjugates may be capable to increase chemotherapy efficacy of anti-cancer drugs that are suboptimal due to the drug resistance.
Collapse
|
31
|
Ganapathi RN, Ganapathi MK. Mechanisms regulating resistance to inhibitors of topoisomerase II. Front Pharmacol 2013; 4:89. [PMID: 23914174 PMCID: PMC3729981 DOI: 10.3389/fphar.2013.00089] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 06/24/2013] [Indexed: 11/13/2022] Open
Abstract
Inhibitors of topoisomerase II (topo II) are clinically effective in the management of hematological malignancies and solid tumors. The efficacy of anti-tumor drugs targeting topo II is often limited by resistance and studies with in vitro cell culture models have provided several insights on potential mechanisms. Multidrug transporters that are involved in the efflux and consequently reduced cytotoxicity of diverse anti-tumor agents suggest that they play an important role in resistance to clinically active drugs. However, in clinical trials, modulating the multidrug-resistant phenotype with agents that inhibit the efflux pump has not had an impact. Since reduced drug accumulation per se is insufficient to explain tumor cell resistance to topo II inhibitors several studies have focused on characterizing mechanisms that impact on DNA damage mediated by drugs that target the enzyme. Mammalian topo IIα and topo IIβ isozymes exhibit similar catalytic, but different biologic, activities. Whereas topo IIα is associated with cell division, topo IIβ is involved in differentiation. In addition to site specific mutations that can affect drug-induced topo II-mediated DNA damage, post-translation modification of topo II primarily by phosphorylation can potentially affect enzyme-mediated DNA damage and the downstream cytotoxic response of drugs targeting topo II. Signaling pathways that can affect phosphorylation and changes in intracellular calcium levels/calcium dependent signaling that can regulate site-specific phosphorylation of topoisomerase have an impact on downstream cytotoxic effects of topo II inhibitors. Overall, tumor cell resistance to inhibitors of topo II is a complex process that is orchestrated not only by cellular pharmacokinetics but more importantly by enzymatic alterations that govern the intrinsic drug sensitivity.
Collapse
Affiliation(s)
- Ram N Ganapathi
- Levine Cancer Institute, Carolinas HealthCare System Charlotte, NC, USA
| | | |
Collapse
|
32
|
Chen J, Ouyang J, Kong J, Zhong W, Xing MM. Photo-cross-linked and pH-sensitive biodegradable micelles for doxorubicin delivery. ACS APPLIED MATERIALS & INTERFACES 2013; 5:3108-17. [PMID: 23530535 DOI: 10.1021/am400017q] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Cross-linked polymeric micelles have gained increasing research interest in the past decade due to the instability of existing polymeric micelles when used in vivo. In this study, we reported a series of covalently cross-linked pH-sensitive biodegradable micelles based on the poly(ethylene glycol)-hyperbranched poly(β-aminoester)s with acrylate group terminals (PEG-HBPAE-A) copolymers for intracellular delivery of doxorubicin (DOX). PEG-HBPAE-A can be self-assembled to form micellar nanoparticles in aqueous solution with diameters of approximately 160 nm. The non-cross-linked micelles (NCLMs) were cross-linked upon UV irradiation to form cross-linked micelles (CLMs). (1)H NMR, FT-IR and dynamic light scattering (DLS) were utilized to investigate the process of the UV cross-linking and the stability of CLMs. The results showed the significantly enhanced stability for CLMs in comparison to NCLMs. pH sensitivity of CLMs and NCLMs were also estimated by DLS. In vitro drug release studies confirmed that DOX release from DOX-loaded CLMs was greatly inhibited upon the neutral pH environment, whereas DOX underwent faster release in acidic conditions. MTT assays showed that DOX-loaded micelles had a similar inhibition rate for HepG-2 and MCF-7 cell lines compared with free DOX, whereas the blank CLMs and NCLMs showed very low cytotoxicity. Laser scanning confocal microscopy and real-time in situ fluorescence microscopy were exploited to investigate drug uptake in cells and drug distribution in the interior of cells. These results showed a promising nanocarrier for intracellular DOX delivery with great potential for cancer therapy.
Collapse
Affiliation(s)
- Jun Chen
- Department of Mechanical and Manufacturing Engineering, University of Manitoba, Winnipeg, Manitoba R3T 2N2, Canada
| | | | | | | | | |
Collapse
|
33
|
Ding F, Cai S, William R, Liu XW. Pathways leading to 3-amino- and 3-nitro-2,3-dideoxy sugars: strategies and synthesis. RSC Adv 2013. [DOI: 10.1039/c3ra40595h] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
34
|
Jin Z, Zong C, Jiang B, Zhou Z, Tong J, Cao Y. The effect of combined exposure of 900 MHz radiofrequency fields and doxorubicin in HL-60 cells. PLoS One 2012; 7:e46102. [PMID: 23029402 PMCID: PMC3460948 DOI: 10.1371/journal.pone.0046102] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 08/28/2012] [Indexed: 11/19/2022] Open
Abstract
Human promyelocytic leukemia HL-60 cells were pre-exposed to non-ionizing 900 MHz radiofrequency fields (RF) at 12 µW/cm2 power density for 1 hour/day for 3 days and then treated with a chemotherapeutic drug, doxorubicin (DOX, 0.125 mg/L). Several end-points related to toxicity, viz., viability, apoptosis, mitochondrial membrane potential (MMP), intracellular free calcium (Ca2+) and Ca2+-Mg2+ -ATPase activity were measured. The results obtained in un-exposed and sham-exposed control cells were compared with those exposed to RF alone, DOX alone and RF+DOX. The results indicated no significant differences between un-exposed, sham-exposed control cells and those exposed to RF alone while treatment with DOX alone showed a significant decrease in viability, increased apoptosis, decreased MMP, increased Ca2+ and decreased Ca2+-Mg2+-ATPase activity. When the latter results were compared with cells exposed RF+DOX, the data showed increased cell proliferation, decreased apoptosis, increased MMP, decreased Ca2+ and increased Ca2+-Mg2+-ATPase activity. Thus, RF pre-exposure appear to protect the HL-60 cells from the toxic effects of subsequent treatment with DOX. These observations were similar to our earlier data which suggested that pre-exposure of mice to 900 MHz RF at 120 µW/cm2 power density for 1 hours/day for 14 days had a protective effect in hematopoietic tissue damage induced by subsequent gamma-irradiation.
Collapse
MESH Headings
- Adenosine Triphosphatases/metabolism
- Antibiotics, Antineoplastic/therapeutic use
- Apoptosis/drug effects
- Apoptosis/radiation effects
- Calcium/metabolism
- Cell Survival/drug effects
- Cell Survival/radiation effects
- Doxorubicin/therapeutic use
- HL-60 Cells
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Leukemia, Promyelocytic, Acute/therapy
- Magnesium/metabolism
- Membrane Potential, Mitochondrial/drug effects
- Membrane Potential, Mitochondrial/radiation effects
- Radiofrequency Therapy
Collapse
Affiliation(s)
| | | | | | | | | | - Yi Cao
- School of Public Health, Soochow University, Suzhou, Jiangsu, People's Republic of China
- * E-mail:
| |
Collapse
|
35
|
Li Y, Zheng X, Cao Z, Xu W, Zhang J, Gong M. Self-assembled peptide (CADY-1) improved the clinical application of doxorubicin. Int J Pharm 2012; 434:209-14. [DOI: 10.1016/j.ijpharm.2012.06.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 05/13/2012] [Accepted: 06/01/2012] [Indexed: 01/21/2023]
|
36
|
Low molecular weight β-glucan stimulates doxorubicin-induced suppression of immune functions in mice. Food Sci Biotechnol 2012. [DOI: 10.1007/s10068-012-0084-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
37
|
Doxorubicin selects for fluconazole-resistant petite mutants in Candida glabrata isolates. Int J Med Microbiol 2012; 302:155-61. [PMID: 22664377 DOI: 10.1016/j.ijmm.2012.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Revised: 04/26/2012] [Accepted: 04/29/2012] [Indexed: 01/10/2023] Open
Abstract
Candida infections are a permanent threat to immunocompromised individuals such as cancer patients, and Candida glabrata has emerged as a major problem in recent years. Resistance may develop during lengthy antifungal therapies and is often mediated by upregulation of fungal drug efflux pumps. During chemotherapy the yeast cell is also exposed to cytotoxic agents that may affect its drug susceptibility. Four C. glabrata isolates, three susceptible and one resistant to fluconazole (FLU), were incubated with 20 μg/ml of doxorubicin (DOX) for 90 min. In a second experiment, the isolates were cultured with DOX for ten days. Samples were taken on subsequent days to determine the minimal inhibitory concentration (MIC) of FLU and to analyze expression of CgCDR1, CgCDR2, CgSNQ2 and CgPDR1. Samples were also used to assess the petite phenotype. Short-term DOX exposure did not induce efflux pump gene expression, but genes were consistently overexpressed in FLU-susceptible isolates during long-term exposure. An increase in MIC values on day 6 in two of the isolates coincided with the first occurrence of petite mutants in all susceptible isolates. The respiratory deficiency of selected petite mutants was confirmed by culturing mutants on agar containing glycerol as the sole carbon source. FLU MIC values for respiratory-deficient clones were ≥64 μg/ml, and efflux pump gene expression was greatly increased. The resistant isolate did not develop mitochondrial dysfunction. In summary, the cytotoxic agent DOX selects for FLU-resistant respiratory-deficient C. glabrata mutants, which may affect antifungal therapy.
Collapse
|
38
|
Stewart A, Huang J, Fisher RA. RGS Proteins in Heart: Brakes on the Vagus. Front Physiol 2012; 3:95. [PMID: 22685433 PMCID: PMC3368389 DOI: 10.3389/fphys.2012.00095] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/27/2012] [Indexed: 12/14/2022] Open
Abstract
It has been nearly a century since Otto Loewi discovered that acetylcholine (ACh) release from the vagus produces bradycardia and reduced cardiac contractility. It is now known that parasympathetic control of the heart is mediated by ACh stimulation of G(i/o)-coupled muscarinic M2 receptors, which directly activate G protein-coupled inwardly rectifying potassium (GIRK) channels via Gβγ resulting in membrane hyperpolarization and inhibition of action potential (AP) firing. However, expression of M2R-GIRK signaling components in heterologous systems failed to recapitulate native channel gating kinetics. The missing link was identified with the discovery of regulator of G protein signaling (RGS) proteins, which act as GTPase-activating proteins to accelerate the intrinsic GTPase activity of Gα resulting in termination of Gα- and Gβγ-mediated signaling to downstream effectors. Studies in mice expressing an RGS-insensitive Gα(i2) mutant (G184S) implicated endogenous RGS proteins as key regulators of parasympathetic signaling in heart. Recently, two RGS proteins have been identified as critical regulators of M2R signaling in heart. RGS6 exhibits a uniquely robust expression in heart, especially in sinoatrial (SAN) and atrioventricular nodal regions. Mice lacking RGS6 exhibit increased bradycardia and inhibition of SAN AP firing in response to CCh as well as a loss of rapid activation and deactivation kinetics and current desensitization for ACh-induced GIRK current (I(KACh)). Similar findings were observed in mice lacking RGS4. Thus, dysregulation in RGS protein expression or function may contribute to pathologies involving aberrant electrical activity in cardiac pacemaker cells. Moreover, RGS6 expression was found to be up-regulated in heart under certain pathological conditions, including doxorubicin treatment, which is known to cause life-threatening cardiotoxicity and atrial fibrillation in cancer patients. On the other hand, increased vagal tone may be cardioprotective in heart failure where acetylcholinesterase inhibitors and vagal stimulation have been proposed as potential therapeutics. Together, these studies identify RGS proteins, especially RGS6, as new therapeutic targets for diseases such as sick sinus syndrome or other maladies involving abnormal autonomic control of the heart.
Collapse
Affiliation(s)
- Adele Stewart
- Department of Pharmacology, Carver College of Medicine, University of Iowa Iowa City, IA, USA
| | | | | |
Collapse
|
39
|
Ratterree W, Gieger T, Pariaut R, Saelinger C, Strickland K. Value of echocardiography and electrocardiography as screening tools prior to Doxorubicin administration. J Am Anim Hosp Assoc 2012; 48:89-96. [PMID: 22267175 DOI: 10.5326/jaaha-ms-5680] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The dose-limiting toxicity of doxorubicin is cardiotoxicosis. The authors of this report hypothesized that by using their institution's adopted guidelines (that involve prescreening echocardiography and electrocardiography), they would detect pre-existing cardiac abnormalities that preclude doxorubicin administration in <10% of dogs. Of 101 dogs, only 6 were excluded from doxorubicin administration based on electrocardiogram abnormalities, with a majority of those arrhythmias classified as ventricular premature contractions. One patient was excluded based on echocardiogram alone due to hypertrophic cardiomyopathy. The incidence of cardiotoxicity in treated dogs was 8% (8/101). Additional pretreatment and ongoing studies are indicated to identify risk factors for cardiotoxicity.
Collapse
Affiliation(s)
- William Ratterree
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, USA.
| | | | | | | | | |
Collapse
|
40
|
Chen J, Zehtabi F, Ouyang J, Kong J, Zhong W, Xing MMQ. Reducible self-assembled micelles for enhanced intracellular delivery of doxorubicin. ACTA ACUST UNITED AC 2012. [DOI: 10.1039/c2jm15277k] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
41
|
Huang J, Yang J, Maity B, Mayuzumi D, Fisher RA. Regulator of G protein signaling 6 mediates doxorubicin-induced ATM and p53 activation by a reactive oxygen species-dependent mechanism. Cancer Res 2011; 71:6310-9. [PMID: 21859827 DOI: 10.1158/0008-5472.can-10-3397] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Doxorubicin (DXR), among the most widely used cancer chemotherapy agents, promotes cancer cell death via activation of ataxia telangiectasia mutated (ATM) and the resultant upregulation of tumor suppressor p53. The exact mechanism by which DXR activates ATM is not fully understood. Here, we discovered a novel role for regulator of G protein signaling 6 (RGS6) in mediating activation of ATM and p53 by DXR. RGS6 was robustly induced by DXR, and genetic loss of RGS6 dramatically impaired DXR-induced activation of ATM and p53, as well as its in vivo apoptotic actions in heart. The ability of RGS6 to promote p53 activation in response to DXR was independent of RGS6 interaction with G proteins but required ATM. RGS6 mediated activation of ATM and p53 by DXR via a reactive oxygen species (ROS)-dependent and DNA damage-independent mechanism. This mechanism represents the primary means by which DXR promotes activation of the ATM-p53 apoptosis pathway that underlies its cytotoxic activity. Our findings contradict the canonical theories that DXR activates ATM primarily by promoting DNA damage either directly or indirectly (via ROS) and that RGS6 function is mediated by its interactions with G proteins. These findings reveal a new mechanism for the chemotherapeutic actions of DXR and identify RGS6 as a novel target for cancer chemotherapy.
Collapse
Affiliation(s)
- Jie Huang
- Department of Pharmacology, University of Iowa, Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
42
|
Smuder AJ, Kavazis AN, Min K, Powers SK. Exercise protects against doxorubicin-induced markers of autophagy signaling in skeletal muscle. J Appl Physiol (1985) 2011; 111:1190-8. [PMID: 21778418 DOI: 10.1152/japplphysiol.00429.2011] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Doxorubicin (DOX) is an effective antitumor agent used in cancer treatment. Unfortunately, DOX is also toxic to skeletal muscle and can result in significant muscle wasting. The cellular mechanism(s) by which DOX induces toxicity in skeletal muscle fibers remains unclear. Nonetheless, DOX-induced toxicity is associated with increased generation of reactive oxygen species, oxidative damage, and activation of the calpain and caspase-3 proteolytic systems within muscle fibers. It is currently unknown if autophagy, a proteolytic system that can be triggered by oxidative stress, is activated in skeletal muscles following DOX treatment. Therefore, we tested the hypothesis that systemic administration of DOX leads to increased expression of autophagy markers in the rat soleus muscle. Our results reveal that DOX administration results in increased muscle mRNA levels and/or protein abundance of several important autophagy proteins, including: Beclin-1, Atg12, Atg7, LC3, LC3II-to-LCI ratio, and cathepsin L. Furthermore, given that endurance exercise increases skeletal muscle antioxidant capacity and protects muscle against DOX-induced oxidative stress, we performed additional experiments to determine whether exercise training before DOX administration would attenuate DOX-induced increases in expression of autophagy genes. Our results clearly show that exercise can protect skeletal muscle from DOX-induced expression of autophagy genes. Collectively, our findings indicate that DOX administration increases the expression of autophagy genes in skeletal muscle, and that exercise can protect skeletal muscle against DOX-induced activation of autophagy.
Collapse
Affiliation(s)
- Ashley J Smuder
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, Florida, USA
| | | | | | | |
Collapse
|
43
|
Phytochemical and cytotoxic investigations of Curcuma mangga rhizomes. Molecules 2011; 16:4539-48. [PMID: 21629182 PMCID: PMC6264423 DOI: 10.3390/molecules16064539] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Revised: 04/18/2011] [Accepted: 04/20/2011] [Indexed: 11/17/2022] Open
Abstract
Investigations on the cytotoxic effects of the crude methanol and fractionated extracts (hexane, ethyl acetate) C. mangga against six human cancer cell lines, namely the hormone-dependent breast cell line (MCF-7), nasopharyngeal epidermoid cell line (KB), lung cell line (A549), cervical cell line (Ca Ski), colon cell lines (HCT 116 and HT-29), and one non-cancer human fibroblast cell line (MRC-5) were conducted using an in-vitro neutral red cytotoxicity assay. The crude methanol and fractionated extracts (hexane and ethyl acetate) displayed good cytotoxic effects against MCF-7, KB, A549, Ca Ski and HT-29 cell lines, but exerted no damage on the MRC-5 line. Chemical investigation from the hexane and ethyl acetate fractions resulted in the isolation of seven pure compounds, namely (E)-labda-8(17),12-dien-15,16-dial (1), (E)-15,16-bisnor-labda-8(17),11-dien-13-on (2), zerumin A (3), β-sitosterol, curcumin, demethoxycurcumin and bis-demethoxycurcumin. Compounds 1 and 3 exhibited high cytotoxic effects against all six selected cancer cell lines, while compounds 2 showed no anti-proliferative activity on the tested cell lines. Compound 1 also demonstrated strong cytotoxicity against the normal cell line MRC-5. This paper reports for the first time the cytotoxic activities of C. mangga extracts on KB, A549, Ca Ski, HT-29 and MRC-5, and the occurrence of compound 2 and 3 in C. mangga.
Collapse
|
44
|
Stereo- and regioselective glycosylation of 4-deoxy-ε-rhodomycinone. Carbohydr Res 2011; 346:858-62. [DOI: 10.1016/j.carres.2011.01.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Revised: 01/11/2011] [Accepted: 01/26/2011] [Indexed: 11/21/2022]
|
45
|
Smuder AJ, Kavazis AN, Min K, Powers SK. Exercise protects against doxorubicin-induced oxidative stress and proteolysis in skeletal muscle. J Appl Physiol (1985) 2011; 110:935-42. [PMID: 21310889 DOI: 10.1152/japplphysiol.00677.2010] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Doxorubicin (Dox) is a potent antitumor agent used in cancer treatment. Unfortunately, Dox is myotoxic and results in significant reductions in skeletal muscle mass and function. Complete knowledge of the mechanism(s) by which Dox induces toxicity in skeletal muscle is incomplete, but it is established that Dox-induced toxicity is associated with increased generation of reactive oxygen species and oxidative damage within muscle fibers. Since muscular exercise promotes the expression of numerous cytoprotective proteins (e.g., antioxidant enzymes, heat shock protein 72), we hypothesized that muscular exercise will attenuate Dox-induced damage in exercise-trained muscle fibers. To test this postulate, Sprague-Dawley rats were randomly assigned to the following groups: sedentary, exercise, sedentary with Dox, or exercise with Dox. Our results show increased oxidative stress and activation of cellular proteases (calpain and caspase-3) in skeletal muscle of animals treated with Dox. Importantly, our findings reveal that exercise can prevent the Dox-induced oxidative damage and protease activation in the trained muscle. This exercise-induced protection against Dox-induced toxicity may be due, at least in part, to an exercise-induced increase in muscle levels of antioxidant enzymes and heat shock protein 72. Together, these novel results demonstrate that muscular exercise is a useful countermeasure that can protect skeletal muscle against Dox treatment-induced oxidative stress and protease activation in skeletal muscles.
Collapse
Affiliation(s)
- Ashley J Smuder
- Dept. of Applied Physiology and Kinesiology, Univ. of Florida, Gainesville, FL 32611, USA.
| | | | | | | |
Collapse
|
46
|
Sayed-Ahmed MM. Role of carnitine in cancer chemotherapy-induced multiple organ toxicity. Saudi Pharm J 2010; 18:195-206. [PMID: 23960728 PMCID: PMC3730973 DOI: 10.1016/j.jsps.2010.07.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 06/29/2010] [Indexed: 01/27/2023] Open
Abstract
In the last few years, cancer chemotherapy has been successfully employed in the treatment of different types of human tumours. Unfortunately, the optimal clinical usefulness of this important treatment modality is usually limited secondary to the development of life-threatening multiple organ toxicity. Cancer chemotherapy may cause these toxic effects by mechanisms not involved in their anticancer activity that can severely affect the life of patients and represent a direct cause of death. Several experimental and clinical studies have demonstrated that some important anticancer drugs interfere with the absorption, synthesis, and excretion of carnitine in non-tumour tissues, resulting in a secondary carnitine deficiency which is reversed by carnitine treatment without affecting anticancer therapeutic efficacy. Prototypes of anticancer drugs that alter carnitine system are doxorubicin, cisplatin, carboplatin, oxaliplatin, cyclophosphamide and ifosfamide. Furthermore, cachectic cancer patients are especially at risk for carnitine deficiency due to decreased oral intake and/or increased renal losses. Altered serum and urine carnitine levels have been reported in cancer patients with various forms of malignant diseases. Recent studies in our laboratory have demonstrated that carnitine deficiency constitute a risk factor and should be viewed as a mechanism during development of oxazaphosphorines-induced cardiotoxicity in rats. Similarly, inhibition of gene expression of heart fatty acid-binding protein and organic cation/carnitine transporter in doxorubicin cardiomyopathic rat model has been reported. In view of these facts and in view of irreplaceability of these important anticancer drugs, this review aimed to highlight the role of carnitine depletion and supplementation during development of chemotherapy-induced multiple organ toxicity.
Collapse
Affiliation(s)
- Mohamed M. Sayed-Ahmed
- Department of Pharmacology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|
47
|
|
48
|
Sayed-Ahmed MM, Al-Shabanah OA, Hafez MM, Aleisa AM, Al-Rejaie SS. Inhibition of gene expression of heart fatty acid binding protein and organic cation/carnitine transporter in doxorubicin cardiomyopathic rat model. Eur J Pharmacol 2010; 640:143-9. [DOI: 10.1016/j.ejphar.2010.05.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Revised: 04/13/2010] [Accepted: 05/03/2010] [Indexed: 11/17/2022]
|
49
|
Cui Y, Piao CS, Ha KC, Kim DS, Lee GH, Kim HK, Chae SW, Lee YC, Park SJ, Yoo WH, Kim HR, Chae HJ. Measuring adriamycin-induced cardiac hemodynamic dysfunction with a proteomics approach. Immunopharmacol Immunotoxicol 2010; 32:376-86. [DOI: 10.3109/08923970903440168] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
50
|
Impact of 1.8-GHz radiofrequency radiation (RFR) on DNA damage and repair induced by doxorubicin in human B-cell lymphoblastoid cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2010; 695:16-21. [DOI: 10.1016/j.mrgentox.2009.10.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 09/14/2009] [Accepted: 10/03/2009] [Indexed: 11/20/2022]
|