1
|
Hubiernatorova A, Novak J, Vaskovicova M, Sekac D, Kropyvko S, Hodny Z. Tristetraprolin affects invasion-associated genes expression and cell motility in triple-negative breast cancer model. Cytoskeleton (Hoboken) 2025; 82:311-326. [PMID: 39319680 PMCID: PMC12063522 DOI: 10.1002/cm.21934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Tristetraprolin (TTP) is an RNA-binding protein that negatively regulates its target mRNAs and has been shown to inhibit tumor progression and invasion. Tumor invasion requires precise regulation of cytoskeletal components, and dysregulation of cytoskeleton-associated genes can significantly alter cell motility and invasive capability. Several genes, including SH3PXD2A, SH3PXD2B, CTTN, WIPF1, and WASL, are crucial components of the cytoskeleton reorganization machinery and are essential for adequate cell motility. These genes are also involved in invasion processes, with SH3PXD2A, SH3PXD2B, WIPF1, and CTTN being key components of invadopodia-specialized structures that facilitate invasion. However, the regulation of these genes is not well understood. This study demonstrates that ectopic expression of TTP in MDA-MB-231 cells leads to decreased mRNA levels of CTTN and SH3PXD2A, as well as defects in cell motility and actin filament organization. Additionally, doxorubicin significantly increases TTP expression and reduces the mRNA levels of cytoskeleton-associated genes, enhancing our understanding of how doxorubicin may affect the transcriptional profile of cells. However, doxorubicin affects target mRNAs differently than TTP ectopic expression, suggesting it may not be the primary mechanism of doxorubicin in breast cancer (BC) treatment. High TTP expression is considered as a positive prognostic marker in multiple cancers, including BC. Given that doxorubicin is a commonly used drug for treating triple-negative BC, using TTP as a prognostic marker in this cohort of patients might be limited since it might be challenging to understand if high TTP expression occurred due to the favorable physiological state of the patient or as a consequence of treatment.
Collapse
Affiliation(s)
- Anastasiia Hubiernatorova
- Department of Functional GenomicsInstitute of Molecular Biology and Genetics NAS of UkraineKyivUkraine
- Laboratory of Cell Regeneration and PlasticityInstitute of Animal Physiology and Genetics of the Czech Academy of SciencesLibechovCzech Republic
| | - Josef Novak
- Laboratory of Genome IntegrityInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Michaela Vaskovicova
- Department of Cell Biology, Faculty of ScienceCharles UniversityPragueCzech Republic
- Laboratory of DNA IntegrityInstitute of Animal Physiology and Genetics of the Czech Academy of SciencesLibechovCzech Republic
| | - David Sekac
- Laboratory of Cell Regeneration and PlasticityInstitute of Animal Physiology and Genetics of the Czech Academy of SciencesLibechovCzech Republic
- Department of Cell Biology, Faculty of ScienceCharles UniversityPragueCzech Republic
| | - Serhii Kropyvko
- Department of Functional GenomicsInstitute of Molecular Biology and Genetics NAS of UkraineKyivUkraine
| | - Zdenek Hodny
- Laboratory of Genome IntegrityInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| |
Collapse
|
2
|
Afriza D, Arma U, Faslah R, Suriyah WH. Anticancer Potential of Quercetin on Oral Squamous Cell Carcinoma: A Scoping Review and Molecular Docking. Eur J Dent 2025; 19:15-23. [PMID: 39348855 PMCID: PMC11750326 DOI: 10.1055/s-0044-1789016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a type of cancer that has a low survival rate and high recurrence and metastasis rates. To date, there is still no effective treatment for OSCC. Various types of cancer, including OSCC, have reported quercetin to act as an anticancer agent, but there is no clear research data on how it may affect OSCC. To determine the anticancer potential of quercetin in OSCC, we conducted a scoping review, and to determine the interaction of quercetin with one of the proteins that plays a role in carcinogenesis, namely, BCL-2, we conducted molecular docking. The scoping review process was conducted based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses Extension for Scoping Reviews. The scoping review was searched by collecting articles related to the research topic in Google Scholar, PubMed, ScienceDirect, Cochrane, and EBSCOhost databases. All of the literature records found during the search were imported into the Mendeley software to remove duplication. Nine studies were generated after the titles and abstracts were reviewed according to the inclusion and exclusion criteria. After the full-text screening, no studies were excluded, leaving nine publications determined to be eligible for inclusion in the scoping review. Quercetin showed effects on inhibiting cancer invasion, migration, proliferation, and many protein expressions, as well as increasing cell apoptosis. Molecular docking was done for quercetin and BCl-2 protein. Doxorubicin was utilized as a comparison ligand. The in silico study was utilized using AutoDock Vina, AutoDock Tools 1.5.6, Biovia Discovery Studio 2021, and PyMol. Molecular docking indicated quercetin has a strong binding affinity with BCl-2 protein (ΔG -7.2 kcal/mol). Both scoping review and molecular docking revealed that quercetin is a promising candidate for anticancer agent.
Collapse
Affiliation(s)
- Dhona Afriza
- Department of Oral Biology, Faculty of Dentistry, Baiturrahmah University, Kota Padang, Sumatera Barat, Indonesia
| | - Utmi Arma
- Department of Oral Medicine, Faculty of Dentistry, Baiturrahmah University, Kota Padang, Sumatera Barat, Indonesia
| | - Raefany Faslah
- Department of Oral Medicine, Faculty of Dentistry, Baiturrahmah University, Kota Padang, Sumatera Barat, Indonesia
| | | |
Collapse
|
3
|
O’Dowling AT, Rodriguez BJ, Gallagher TK, Thorpe SD. Machine learning and artificial intelligence: Enabling the clinical translation of atomic force microscopy-based biomarkers for cancer diagnosis. Comput Struct Biotechnol J 2024; 24:661-671. [PMID: 39525667 PMCID: PMC11543504 DOI: 10.1016/j.csbj.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024] Open
Abstract
The influence of biomechanics on cell function has become increasingly defined over recent years. Biomechanical changes are known to affect oncogenesis; however, these effects are not yet fully understood. Atomic force microscopy (AFM) is the gold standard method for measuring tissue mechanics on the micro- or nano-scale. Due to its complexity, however, AFM has yet to become integrated in routine clinical diagnosis. Artificial intelligence (AI) and machine learning (ML) have the potential to make AFM more accessible, principally through automation of analysis. In this review, AFM and its use for the assessment of cell and tissue mechanics in cancer is described. Research relating to the application of artificial intelligence and machine learning in the analysis of AFM topography and force spectroscopy of cancer tissue and cells are reviewed. The application of machine learning and artificial intelligence to AFM has the potential to enable the widespread use of nanoscale morphologic and biomechanical features as diagnostic and prognostic biomarkers in cancer treatment.
Collapse
Affiliation(s)
- Aidan T. O’Dowling
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- Department of Hepatobiliary and Transplant Surgery, St Vincent’s University Hospital, Dublin, Ireland
| | - Brian J. Rodriguez
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- UCD School of Physics, University College Dublin, Dublin, Ireland
| | - Tom K. Gallagher
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- Department of Hepatobiliary and Transplant Surgery, St Vincent’s University Hospital, Dublin, Ireland
| | - Stephen D. Thorpe
- UCD School of Medicine, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- Trinity Centre for Bioengineering, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
4
|
Luo HY, Jiang C, Dou SX, Wang PY, Li H. Quantum Dot-Based Three-Dimensional Single-Particle Tracking Characterizes the Evolution of Spatiotemporal Heterogeneity in Necrotic Cells. Anal Chem 2024; 96:11682-11689. [PMID: 38979688 DOI: 10.1021/acs.analchem.4c00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Cell death is a fundamental biological process with different modes including apoptosis and necrosis. In contrast to programmed apoptosis, necrosis was previously considered disordered and passive, but it is now being realized to be under regulation by certain biological pathways. However, the intracellular dynamics that coordinates with cellular structure changes during necrosis remains unknown, limiting our understanding of the principles of necrosis. Here, we characterized the spatiotemporal intracellular diffusion dynamics in cells undergoing necrosis, using three-dimensional single-particle tracking of quantum dots. We found temporally increased diffusion rates in necrotic cells and spatially enhanced diffusion heterogeneity in the cell periphery, which could be attributed to the reduced molecular crowding resulting from cell swelling and peripheral blebbing, respectively. Moreover, the three-dimensional intracellular diffusion transits from strong anisotropy to nearly isotropy, suggesting a remodeling of the cytoarchitecture that relieves the axial constraint on intracellular diffusion during necrosis. Our results reveal the remarkable alterations of intracellular diffusion dynamics and biophysical properties in necrosis, providing insight into the well-organized nonequilibrium necrotic cell death from a biophysical perspective.
Collapse
Affiliation(s)
- Hong-Yu Luo
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chao Jiang
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuo-Xing Dou
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng-Ye Wang
- Beijing National Laboratory for Condensed Matter Physics and Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100190, China
- School of Physical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Songshan Lake Materials Laboratory, Dongguan, Guangdong 523808, China
| | - Hui Li
- School of Systems Science and Institute of Nonequilibrium Systems, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
5
|
Üstüner E, Yıldırım E, Macun HC, Ekici H, Şahin Y, Güncüm E, Anteplioğlu T, Elifoğlu TB, Bozkaya E. Ultrasonographic and histopathological investigation of the effect of N-acetylcysteine on doxorubicin-induced ovarian and uterine toxicity in rats. J Ovarian Res 2024; 17:135. [PMID: 38943148 PMCID: PMC11214216 DOI: 10.1186/s13048-024-01459-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/17/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND This study aimed to investigate the mitigating effect of N-acetylcysteine (NAC) on doxorubicin (DOX)-induced ovarian and uterine toxicity in rats using laboratory tests, ultrasonographic (US) imaging, and histopathology analysis. METHODS Forty-eight rats were divided into six groups (n = 8) as follows: Group A (control) (0.5 mL saline administered intraperitoneally [IP]), Group B (a single 10 mg/kg dose of DOX administered IP on day 1), Group C (a single 10 mg/kg dose of DOX administered IP 24 h before sacrifice), Group D (100 mg/kg of NAC administered IP for 21 days), Group E ( a single 10 mg/kg dose of DOX administered IP on day 1 and 100 mg/kg of NAC administered IP for 21 days), and Group F (100 mg/kg of NAC administered IP for 21 days and a single 10 mg/kg dose of DOX administered IP 24 h before sacrifice). The ovaries were examined using B-mode US on days 1, 14, and 21, and the histopathological examinations of the ovaries and the uterus were undertaken after sacrifice on day 22. RESULTS Histomorphological analyses showed that ovarian weight decreased after DOX administration in Group B but not in Group E. US revealed a transient increase in ovarian size in Group B and E, reverting to baseline levels over time, as well as a progressive increase in peritoneal fluid in Groups B and E. Group B exhibited a significant decrease in the thickness of the endometrium and myometrium and uterine cornual length, which was not observed in Group E. Histopathological examination showed that DOX caused a decline in follicular count, especially in primordial, secondary, and Graafian follicles, and resulted in follicular atresia, predominantly in Group B. Destructive degeneration/necrosis and vascular changes were most prominently seen in the corpus luteum of Groups C and B. In NAC-treated rats (Groups E and F), although germ cell damage was present, atretic follicles and vascular changes, such as hyperemia and congestion, were reduced. The anti-müllerian hormone (AMH) level was the highest in Group F. CONCLUSIONS NAC, an antioxidant, attenuated DOX-induced gonadotoxicity in rats.
Collapse
Affiliation(s)
- Evren Üstüner
- Faculty of Medicine, Department of Radiology, Ankara University, Ankara, Turkey.
| | - Ebru Yıldırım
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Kirikkale University, Kirikkale, Turkey
| | - Hasan Ceyhun Macun
- Faculty of Veterinary Medicine, Department of Obstetrics and Gynecology, Kirikkale University, Kirikkale, Turkey
| | - Hüsamettin Ekici
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Kirikkale University, Kirikkale, Turkey
| | - Yaşar Şahin
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Kirikkale University, Kirikkale, Turkey
| | - Enes Güncüm
- Faculty of Veterinary Medicine, Department of Pharmacology and Toxicology, Kirikkale University, Kirikkale, Turkey
| | - Tuğçe Anteplioğlu
- Faculty of Veterinary Medicine, Department of Pathology, Kirikkale University, Kirikkale, Turkey
| | - Taha Burak Elifoğlu
- Faculty of Veterinary Medicine, Department of Obstetrics and Gynecology, Kirikkale University, Kirikkale, Turkey
| | - Esra Bozkaya
- Scientific and Technological Research Application and Research Center, Kirikkale University, Kirikkale, Türkiye
| |
Collapse
|
6
|
Yan T, Yu H, Li T, Dong Y. Mechanisms of Cardiovascular Toxicities Induced by Cancer Therapies and Promising Biomarkers for Their Prediction: A Scoping Review. Heart Lung Circ 2024; 33:605-638. [PMID: 38242833 DOI: 10.1016/j.hlc.2023.12.006] [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: 05/31/2023] [Revised: 10/16/2023] [Accepted: 12/01/2023] [Indexed: 01/21/2024]
Abstract
AIM With the advancement of anti-cancer medicine, cardiovascular toxicities due to cancer therapies are common in oncology patients, resulting in increased mortality and economic burden. Cardiovascular toxicities caused by cancer therapies include different severities of cardiomyopathy, arrhythmia, myocardial ischaemia, hypertension, and thrombosis, which may lead to left ventricular dysfunction and heart failure. This scoping review aimed to summarise the mechanisms of cardiovascular toxicities following various anti-cancer treatments and potential predictive biomarkers for early detection. METHODS PubMed, Cochrane, Embase, Web of Science, Scopus, and CINAHL databases were searched for original studies written in English related to the mechanisms of cardiovascular toxicity induced by anti-cancer therapies, including chemotherapy, targeted therapy, immunotherapy, radiation therapy, and relevant biomarkers. The search and title/abstract screening were conducted independently by two reviewers, and the final analysed full texts achieved the consensus of the two reviewers. RESULTS A total of 240 studies were identified based on their titles and abstracts. In total, 107 full-text articles were included in the analysis. Cardiomyocyte and endothelial cell apoptosis caused by oxidative stress injury, activation of cell apoptosis, blocking of normal cardiovascular protection signalling pathways, overactivation of immune cells, and myocardial remodelling were the main mechanisms. Promising biomarkers for anti-cancer therapies related to cardiovascular toxicity included placental growth factor, microRNAs, galectin-3, and myeloperoxidase for the early detection of cardiovascular toxicity. CONCLUSION Understanding the mechanisms of cardiovascular toxicity following various anti-cancer treatments could provide implications for future personalised treatment methods to protect cardiovascular function. Furthermore, specific early sensitive and stable biomarkers of cardiovascular system damage need to be identified to predict reversible damage to the cardiovascular system and improve the effects of anti-cancer agents.
Collapse
Affiliation(s)
- Tingting Yan
- Nursing Department, Liaocheng Vocational and Technical College, Liaocheng City, Shandong Province, China
| | - Hailong Yu
- Department of Gastrointestinal Surgery, Liaocheng People's Hospital, Liaocheng City, Shandong Province, China
| | - Tai Li
- Nursing Department, Liaocheng Vocational and Technical College, Liaocheng City, Shandong Province, China
| | - Yanhong Dong
- Alice Lee Centre for Nursing Studies, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
7
|
Guven C, Taskin E, Aydın Ö, Kaya ST, Sevgiler Y. Diazoxide attenuates DOX-induced cardiotoxicity in cultured rat myocytes. Biotech Histochem 2024; 99:113-124. [PMID: 38439686 DOI: 10.1080/10520295.2024.2324368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Doxorubicin (DOX)-induced cardiotoxicity is a well known clinical problem, and many investigations have been made of its possible amelioration. We have investigated whether diazoxide (DIA), an agonist at mitochondrial ATP-sensitive potassium channels (mitoKATP), could reverse DOX-induced apoptotic myocardial cell loss, in cultured rat cardiomyocytes. The role of certain proteins in this pathway was also studied. The rat cardiomyocyte cell line (H9c2) was treated with DOX, and also co-treated with DOX and DIA, for 24 h. Distribution of actin filaments, mitochondrial membrane potential, superoxide dismutase (SOD) activity, total oxidant and antioxidant status (TOS and TAS, respectively), and some protein expressions, were assessed. DOX significantly decreased SOD activity, increased ERK1/2 protein levels, and depolarised the mitochondrial membrane, while DIA co-treatment inhibited such changes. DIA co-treatment ameliorated DOX-induced cytoskeletal changes via F-actin distribution and mitoKATP structure. Co-treatment also decreased ERK1/2 and cytochrome c protein levels. Cardiomyocyte loss due to oxidative stress-mediated apoptosis is a key event in DOX-induced cytotoxicity. DIA had protective effects on DOX-induced cardiotoxicity, via mitoKATP integrity, especially with elevated SUR2A levels; but also by a cascade including SOD/AMPK/ERK1/2. Therefore, DIA may be considered a candidate agent for protecting cardiomyocytes against DOX chemotherapy.
Collapse
Affiliation(s)
- Celal Guven
- Department of Biophysics, Faculty of Medicine, Adıyaman University, Adıyaman, Turkey
| | - Eylem Taskin
- Department of Physiology, Faculty of Medicine, Adıyaman University, Adıyaman, Turkey
| | - Özgül Aydın
- Department of Biology, Institute of Natural and Applied Sciences, Adıyaman University, Adıyaman, Turkey
| | - Salih Tunç Kaya
- Department of Biology, Faculty of Science and Letters, Düzce University, Düzce, Turkey
| | - Yusuf Sevgiler
- Department of Biology, Faculty of Science and Letters, Adıyaman University, Adıyaman, Turkey
| |
Collapse
|
8
|
Shi J, Wei L. ROCK1 deficiency preserves caveolar compartmentalization of signaling molecules and cell membrane integrity. FASEB Bioadv 2024; 6:85-102. [PMID: 38463696 PMCID: PMC10918988 DOI: 10.1096/fba.2024-00015] [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] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
In this study, we investigated the roles of ROCK1 in regulating structural and functional features of caveolae located at the cell membrane of cardiomyocytes, adipocytes, and mouse embryonic fibroblasts (MEFs) as well as related physiopathological effects. Caveolae are small bulb-shaped cell membrane invaginations, and their roles have been associated with disease conditions. One of the unique features of caveolae is that they are physically linked to the actin cytoskeleton that is well known to be regulated by RhoA/ROCKs pathway. In cardiomyocytes, we observed that ROCK1 deficiency is coincident with an increased caveolar density, clusters, and caveolar proteins including caveolin-1 and -3. In the mouse cardiomyopathy model with transgenic overexpressing Gαq in myocardium, we demonstrated the reduced caveolar density at cell membrane and reduced caveolar protein contents. Interestingly, coexisting ROCK1 deficiency in cardiomyocytes can rescue these defects and preserve caveolar compartmentalization of β-adrenergic signaling molecules including β1-adrenergic receptor and type V/VI adenylyl cyclase. In cardiomyocytes and adipocytes, we detected that ROCK1 deficiency increased insulin signaling with increased insulin receptor activation in caveolae. In MEFs, we identified that ROCK1 deficiency increased caveolar and total levels of caveolin-1 and cell membrane repair ability after mechanical or chemical disruptions. Together, these results demonstrate that ROCK1 can regulate caveolae plasticity and multiple functions including compartmentalization of signaling molecules and cell membrane repair following membrane disruption by mechanical force and oxidative damage. These findings provide possible molecular insights into the beneficial effects of ROCK1 deletion/inhibition in cardiomyocytes, adipocytes, and MEFs under certain diseased conditions.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| |
Collapse
|
9
|
Oraiopoulou ME, Tzamali E, Psycharakis SE, Tzedakis G, Makatounakis T, Manolitsi K, Drakos E, Vakis AF, Zacharakis G, Papamatheakis J, Sakkalis V. The Temozolomide-Doxorubicin paradox in Glioblastoma in vitro-in silico preclinical drug-screening. Sci Rep 2024; 14:3759. [PMID: 38355655 PMCID: PMC10866941 DOI: 10.1038/s41598-024-53684-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: 07/25/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
Adjuvant Temozolomide is considered the front-line Glioblastoma chemotherapeutic treatment; yet not all patients respond. Latest trends in clinical trials usually refer to Doxorubicin; yet it can lead to severe side-effects if administered in high doses. While Glioblastoma prognosis remains poor, little is known about the combination of the two chemotherapeutics. Patient-derived spheroids were generated and treated with a range of Temozolomide/Doxorubicin concentrations either as monotherapy or in combination. Optical microscopy was used to monitor the growth pattern and cell death. Based on the monotherapy experiments, we developed a probabilistic mathematical framework in order to describe the drug-induced effect at the single-cell level and simulate drug doses in combination assuming probabilistic independence. Doxorubicin was found to be effective in doses even four orders of magnitude less than Temozolomide in monotherapy. The combination therapy doses tested in vitro were able to lead to irreversible growth inhibition at doses where monotherapy resulted in relapse. In our simulations, we assumed both drugs are anti-mitotic; Temozolomide has a growth-arrest effect, while Doxorubicin is able to cumulatively cause necrosis. Interestingly, under no mechanistic synergy assumption, the in silico predictions underestimate the in vitro results. In silico models allow the exploration of a variety of potential underlying hypotheses. The simulated-biological discrepancy at certain doses indicates a supra-additive response when both drugs are combined. Our results suggest a Temozolomide-Doxorubicin dual chemotherapeutic scheme to both disable proliferation and increase cytotoxicity against Glioblastoma.
Collapse
Affiliation(s)
- Mariam-Eleni Oraiopoulou
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
- Cancer Research UK - Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Eleftheria Tzamali
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Stylianos E Psycharakis
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
- School of Medicine, University of Crete, Heraklion, Greece
| | - Georgios Tzedakis
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece
| | - Takis Makatounakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
| | - Katina Manolitsi
- University General Hospital of Heraklion (PAGNI), Heraklion, Greece
| | - Elias Drakos
- School of Medicine, University of Crete, Heraklion, Greece
- University General Hospital of Heraklion (PAGNI), Heraklion, Greece
| | - Antonis F Vakis
- School of Medicine, University of Crete, Heraklion, Greece
- University General Hospital of Heraklion (PAGNI), Heraklion, Greece
| | - Giannis Zacharakis
- Institute of Electronic Structure and Laser (IESL), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
| | - Joseph Papamatheakis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology Hellas (FORTH), Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | - Vangelis Sakkalis
- Institute of Computer Science (ICS), Foundation for Research and Technology-Hellas (FORTH), Heraklion, Greece.
| |
Collapse
|
10
|
Alhowail AH, Eggert M, Bloemer J, Pinky PD, Woodie L, Bhattacharya S, Bhattacharya D, Buabeid MA, Smith B, Dhanasekaran M, Piazza G, Reed MN, Escobar M, Arnold RD, Suppiramaniam V. Phenyl-2-aminoethyl selenide ameliorates hippocampal long-term potentiation and cognitive deficits following doxorubicin treatment. PLoS One 2023; 18:e0294280. [PMID: 37948406 PMCID: PMC10637675 DOI: 10.1371/journal.pone.0294280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
Chemotherapy-induced memory loss ("chemobrain") can occur following treatment with the widely used chemotherapeutic agent doxorubicin (DOX). However, the mechanisms through which DOX induces cognitive dysfunction are not clear, and there are no commercially available therapies for its treatment or prevention. Therefore, the aim of this study was to determine the therapeutic potential of phenyl-2-aminoethyl selenide (PAESe), an antioxidant drug previously demonstrated to reduce cardiotoxicity associated with DOX treatment, against DOX-induced chemobrain. Four groups of male athymic NCr nude (nu/nu) mice received five weekly tail-vein injections of saline (Control group), 5 mg/kg of DOX (DOX group), 10 mg/kg PAESe (PAESe group), or 5 mg/kg DOX and 10 mg/kg PAESe (DOX+PAESe group). Spatial memory was evaluated using Y-maze and novel object location tasks, while synaptic plasticity was assessed through the measurement of field excitatory postsynaptic potentials from the Schaffer collateral circuit. Western blot analyses were performed to assess hippocampal protein and phosphorylation levels. In this model, DOX impaired synaptic plasticity and memory, and increased phosphorylation of protein kinase B (Akt) and extracellular-regulated kinase (ERK). Co-administration of PAESe reduced Akt and ERK phosphorylation and ameliorated the synaptic and memory deficits associated with DOX treatment.
Collapse
Affiliation(s)
- Ahmad H. Alhowail
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Kingdom of Saudi Arabia
| | - Matthew Eggert
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Jenna Bloemer
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Priyanka D. Pinky
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Lauren Woodie
- Department of Nutrition, Dietetics and Hospitality Management, College of Human Sciences, Auburn University, Auburn, AL, United States of America
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Dwipayan Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Manal A. Buabeid
- College of Pharmacy and Health Sciences, Ajman University, Ajman, UAE
| | - Bruce Smith
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Muralikrishnan Dhanasekaran
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
| | - Gary Piazza
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
| | - Miranda N. Reed
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
| | - Martha Escobar
- Department of Psychology, Oakland University, Rochester, MI, United States of America
| | - Robert D. Arnold
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
| | - Vishnu Suppiramaniam
- Department of Drug Discovery and Development, Auburn University, Auburn, AL, United States of America
- Center for Neuroscience Initiative, Auburn University, Auburn, AL, United States of America
- Department of Molecular and Cellular Biology, College of Science and Mathematics, Kennesaw State University, Kennesaw, Georgia
| |
Collapse
|
11
|
Crovella S, Ouhtit A, Rahman SM, Rahman MM. Docosahexaenoic Acid, a Key Compound for Enhancing Sensitization to Drug in Doxorubicin-Resistant MCF-7 Cell Line. Nutrients 2023; 15:nu15071658. [PMID: 37049499 PMCID: PMC10097357 DOI: 10.3390/nu15071658] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
Drug resistance is a well-known and significant obstacle in the battle against cancer, rendering chemotherapy treatments often ineffective. To improve the effectiveness of chemotherapy, researchers are exploring the use of natural molecules that can enhance its ability to kill cancer cells and limit their spread. Docosahexaenoic acid (DHA), a lipid found in marine fish, has been shown to enhance the cytotoxicity of various anti-cancer drugs in vitro and in vivo. While the combined use of chemotherapeutic drugs with DHA demonstrated promising preliminary results in clinical trials, there is still a significant amount of information to be discovered regarding the precise mechanism of action of DHA. As the biological pathways involved in the chemosensitization of already chemoresistant MCF-7 cells are still not entirely unraveled, in this study, we aimed to investigate whether DHA co-treatment could enhance the ability of the chemotherapy drug doxorubicin to inhibit the growth and invasion of MCF-7 breast cancer cells (MCF-7/Dox) that had become resistant to the drug. Upon treating MCF-7/Dox cells with DHA or DHA-doxorubicin, it was observed that the DHA-doxorubicin combination effectively enhanced cancer cell death by impeding in vitro propagation and invasive ability. In addition, it led to an increase in doxorubicin accumulation and triggered apoptosis by arresting the cell cycle at the G2/M phase. Other observed effects included a decrease in the multi-drug resistance (MDR) carrier P-glycoprotein (P-gp) and TG2, a tumor survival factor. Augmented quantities of molecules promoting apoptosis such as Bak1 and caspase-3 and enhanced lipid peroxidation were also detected. Our findings in the cell model suggest that DHA can be further investigated as a natural compound to be used alongside doxorubicin in the treatment of breast cancer that is unresponsive to chemotherapy.
Collapse
Affiliation(s)
- Sergio Crovella
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha 2713, Qatar
| | - Allal Ouhtit
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha 2713, Qatar
| | - Shaikh Mizanoor Rahman
- Obesity and Cancer Biology Lab, Natural & Medical Sciences Research Center, University of Nizwa, Nizwa 616, Oman
| | - Md Mizanur Rahman
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha 2713, Qatar
| |
Collapse
|
12
|
Motiei M, Mišík O, Truong TH, Lizal F, Humpolíček P, Sedlařík V, Sáha P. Engineering of inhalable nano-in-microparticles for co-delivery of small molecules and miRNAs. DISCOVER NANO 2023; 18:38. [PMID: 37382704 DOI: 10.1186/s11671-023-03781-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/27/2023] [Indexed: 06/30/2023]
Abstract
In this study, novel Trojan particles were engineered for direct delivery of doxorubicin (DOX) and miR-34a as model drugs to the lungs to raise local drug concentration, decrease pulmonary clearance, increase lung drug deposition, reduce systemic side effects, and overcome multi-drug resistance. For this purpose, targeted polyelectrolyte nanoparticles (tPENs) developed with layer-by-layer polymers (i.e., chitosan, dextran sulfate, and mannose-g-polyethyleneimine) were spray dried into a multiple-excipient (i.e., chitosan, leucine, and mannitol). The resulting nanoparticles were first characterized in terms of size, morphology, in vitro DOX release, cellular internalization, and in vitro cytotoxicity. tPENs showed comparable cellular uptake levels to PENs in A549 cells and no significant cytotoxicity on their metabolic activity. Co-loaded DOX/miR-34a showed a greater cytotoxicity effect than DOX-loaded tPENs and free drugs, which was confirmed by Actin staining. Thereafter, nano-in-microparticles were studied through size, morphology, aerosolization efficiency, residual moisture content, and in vitro DOX release. It was demonstrated that tPENs were successfully incorporated into microspheres with adequate emitted dose and fine particle fraction but low mass median aerodynamic diameter for deposition into the deep lung. The dry powder formulations also demonstrated a sustained DOX release at both pH values of 6.8 and 7.4.
Collapse
Affiliation(s)
- Marjan Motiei
- Centre of Polymer Systems, University Institute, TBU, Tr. Tomase Bati, 5678, Zlin, Czech Republic.
| | - Ondrej Mišík
- Faculty of Mechanical Engineering, Brno University of Technology, Technicka 2896/2, 61669, Brno, Czech Republic
| | - Thanh Huong Truong
- Centre of Polymer Systems, University Institute, TBU, Tr. Tomase Bati, 5678, Zlin, Czech Republic
| | - Frantisek Lizal
- Faculty of Mechanical Engineering, Brno University of Technology, Technicka 2896/2, 61669, Brno, Czech Republic
| | - Petr Humpolíček
- Centre of Polymer Systems, University Institute, TBU, Tr. Tomase Bati, 5678, Zlin, Czech Republic
| | - Vladimír Sedlařík
- Centre of Polymer Systems, University Institute, TBU, Tr. Tomase Bati, 5678, Zlin, Czech Republic
| | - Petr Sáha
- Centre of Polymer Systems, University Institute, TBU, Tr. Tomase Bati, 5678, Zlin, Czech Republic
| |
Collapse
|
13
|
Ganesan R, Prabhakaran VS, Valsala Gopalakrishnan A. Metabolomic Signatures in Doxorubicin-Induced Metabolites Characterization, Metabolic Inhibition, and Signaling Pathway Mechanisms in Colon Cancer HCT116 Cells. Metabolites 2022; 12:1047. [PMID: 36355130 PMCID: PMC9694538 DOI: 10.3390/metabo12111047] [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: 09/28/2022] [Revised: 10/19/2022] [Accepted: 10/28/2022] [Indexed: 01/04/2025] Open
Abstract
Doxorubicin (DOX) is a chemotherapeutic agent is used for various cancer cells. To characterize the chemical structural components and metabolic inhibition, we applied a DOX to HCT116 colon cancer cells using an independent metabolites profiling approach. Chemical metabolomics has been involved in the new drug delivery systems. Metabolomics profiling of DOX-applied HCT116 colon cancer cellular metabolisms is rare. We used 1H nuclear magnetic resonance (NMR) spectroscopy in this study to clarify how DOX exposure affected HCT116 colon cancer cells. Metabolomics profiling in HCT116 cells detects 50 metabolites. Tracking metabolites can reveal pathway activities. HCT116 colon cancer cells were evenly treated with different concentrations of DOX for 24 h. The endogenous metabolites were identified by comparison with healthy cells. We found that acetate, glucose, glutamate, glutamine, sn-glycero-3-phosphocholine, valine, methionine, and isoleucine were increased. Metabolic expression of alanine, choline, fumarate, taurine, o-phosphocholine, inosine, lysine, and phenylalanine was decreased in HCT116 cancer cells. The metabolic phenotypic expression is markedly altered during a high dose of DOX. It is the first time that there is a metabolite pool and phenotypic expression in colon cancer cells. Targeting the DOX-metabolite axis may be a novel strategy for improving the curative effect of DOX-based therapy for colon cancer cells. These methods facilitate the routine metabolomic analysis of cancer cells.
Collapse
Affiliation(s)
- Raja Ganesan
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24253, Korea
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea
| | - Vasantha-Srinivasan Prabhakaran
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, India
| |
Collapse
|
14
|
Gupta P, Makkar TK, Goel L, Pahuja M. Role of inflammation and oxidative stress in chemotherapy-induced neurotoxicity. Immunol Res 2022; 70:725-741. [PMID: 35859244 DOI: 10.1007/s12026-022-09307-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/08/2022] [Indexed: 11/28/2022]
Abstract
Chemotherapeutic agents may adversely affect the nervous system, including the neural precursor cells as well as the white matter. Although the mechanisms are not completely understood, several hypotheses connecting inflammation and oxidative stress with neurotoxicity are now emerging. The proposed mechanisms differ depending on the class of drug. For example, toxicity due to cisplatin occurs due to activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), which alters hippocampal long-term potentiation. Free radical injury is also involved in the cisplatin-mediated neurotoxicity as dysregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) has been seen which protects against the free radical injury by regulating glutathione S-transferases and hemeoxygenase-1 (HO-1). Thus, correcting the imbalance between NF-κB and Nrf2/HO-1 pathways may alleviate cisplatin-induced neurotoxicity. With newer agents like bortezomib, peripheral neuropathy occurs due to up-regulation of TNF-α and IL-6 in the sensory neurons. Superoxide dismutase dysregulation is also involved in bortezomib-induced neuropathy. This article reviews the available literature on inflammation and oxidative stress in neurotoxicity caused by various classes of chemotherapeutic agents. It covers the conventional medicines like platinum compounds, vinca alkaloids, and methotrexate, as well as the newer therapeutic agents like immunomodulators and immune checkpoint inhibitors. A better understanding of the pathophysiology will lead to further advancement in strategies for management of chemotherapy-induced neurotoxicity.
Collapse
Affiliation(s)
- Pooja Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India. .,Coordinator, AIIMS Adverse Drug Reaction Monitoring Centre, Pharmacovigilance Program of India, New Delhi, India.
| | - Tavneet Kaur Makkar
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Lavisha Goel
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Monika Pahuja
- Division of Basic Medical Sciences, Indian Council of Medical Research, New Delhi, India
| |
Collapse
|
15
|
Shi J, Wei L. Rho Kinases in Embryonic Development and Stem Cell Research. Arch Immunol Ther Exp (Warsz) 2022; 70:4. [PMID: 35043239 PMCID: PMC8766376 DOI: 10.1007/s00005-022-00642-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
The Rho-associated coiled-coil containing kinases (ROCKs or Rho kinases) belong to the AGC (PKA/PKG/PKC) family of serine/threonine kinases and are major downstream effectors of small GTPase RhoA, a key regulator of actin-cytoskeleton reorganization. The ROCK family contains two members, ROCK1 and ROCK2, which share 65% overall identity and 92% identity in kinase domain. ROCK1 and ROCK2 were assumed to be functionally redundant, based largely on their major common activators, their high degree kinase domain homology, and study results from overexpression with kinase constructs or chemical inhibitors. ROCK signaling research has expanded to all areas of biology and medicine since its discovery in 1996. The rapid advance is befitting ROCK’s versatile functions in modulating various cell behavior, such as contraction, adhesion, migration, proliferation, polarity, cytokinesis, and differentiation. The rapid advance is noticeably driven by an extensive linking with clinical medicine, including cardiovascular abnormalities, aberrant immune responsive, and cancer development and metastasis. The rapid advance during the past decade is further powered by novel biotechnologies including CRISPR-Cas and single cell omics. Current consensus, derived mainly from gene targeting and RNA interference approaches, is that the two ROCK isoforms have overlapping and distinct cellular, physiological and pathophysiology roles. In this review, we present an overview of the milestone discoveries in ROCK research. We then focus on the current understanding of ROCK signaling in embryonic development, current research status using knockout and knockin mouse models, and stem cell research.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|
16
|
Wardhani BWK, Louisa M, Watanabe Y, Setiabudy R, Kato M. TGF-β-Induced TMEPAI Promotes Epithelial-Mesenchymal Transition in Doxorubicin-Treated Triple-Negative Breast Cancer Cells via SMAD3 and PI3K/AKT Pathway Alteration. BREAST CANCER-TARGETS AND THERAPY 2021; 13:529-538. [PMID: 34584450 PMCID: PMC8464328 DOI: 10.2147/bctt.s325429] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 09/03/2021] [Indexed: 12/27/2022]
Abstract
Introduction Epithelial–mesenchymal transition (EMT) and overexpression of drug efflux transporters have been reported to cause doxorubicin resistance. Our previous study indicated that TMEPAI (transmembrane prostate androgen-induced protein) attenuated doxorubicin sensitivity in triple-negative breast cancer cells. However, how TMEPAI contributes to doxorubicin resistance in TNBC remains unclear. Thus, the present study aimed to elucidate the mechanism of TMEPAI in doxorubicin resistance in triple-negative breast cancer cells. Methods We used BT549, triple-negative cells wild type (WT), and BT549 TMEPAI knock-out. Both cells were treated with TGF-β 2 ng/mL for 24 hours, followed by TGF-β 2 ng/mL and doxorubicin 12.9 nM for another 24 hours. Afterward, the cells were harvested and counted. Cells were further lysed and used for RT-PCR and Western blot analysis. We determined the expression levels of proliferation, apoptosis, EMT markers, and drug efflux transporters. Additionally, we investigated the expressions of PI3K as well as SMAD3 and AKT phosphorylation. Results TNBC cells were shown to be less sensitive to doxorubicin in the presence of TMEPAI. TMEPAI was shown to alleviate the mRNA expressions of apoptosis markers: Bax, Bcl2, Caspase-3, and Caspase-9. Our results indicated that the presence of TMEPAI greatly amplifies EMT and increases drug efflux transporter expressions after doxorubicin treatment. Furthermore, our findings demonstrated that TMEPAI reduced the action of doxorubicin in inhibiting SMAD3 phosphorylation. TMEPAI was also shown to modify the effect of doxorubicin by reducing PI3K expressions and Akt phosphorylation in triple-negative breast cancer cells. Conclusion Our findings indicate that TMEPAI promotes EMT and drug efflux transporters at least in part by shifting doxorubicin action from SMAD3 phosphorylation reduction to PI3K/AKT inhibition in triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Bantari W K Wardhani
- Biomedical Sciences, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia.,Department of Pharmacology, Faculty of Military Pharmacy, Indonesia Defense University, West Java, Indonesia
| | - Melva Louisa
- Department of Pharmacology and Therapeutics, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Yukihide Watanabe
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Rianto Setiabudy
- Department of Pharmacology and Therapeutics, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Mitsuyasu Kato
- Department of Experimental Pathology, Graduate School of Comprehensive Human Sciences, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
17
|
Virtual Screening for Biomimetic Anti-Cancer Peptides from Cordyceps militaris Putative Pepsinized Peptidome and Validation on Colon Cancer Cell Line. Molecules 2021; 26:molecules26195767. [PMID: 34641308 PMCID: PMC8510206 DOI: 10.3390/molecules26195767] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/18/2021] [Accepted: 09/19/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer is one of the leading causes of cancer-related death in Thailand and many other countries. The standard practice for curing this cancer is surgery with an adjuvant chemotherapy treatment. However, the unfavorable side effects of chemotherapeutic drugs are undeniable. Recently, protein hydrolysates and anticancer peptides have become popular alternative options for colon cancer treatment. Therefore, we aimed to screen and select the anticancer peptide candidates from the in silico pepsin hydrolysate of a Cordyceps militaris (CM) proteome using machine-learning-based prediction servers for anticancer prediction, i.e., AntiCP, iACP, and MLACP. The selected CM-anticancer peptide candidates could be an alternative treatment or co-treatment agent for colorectal cancer, reducing the use of chemotherapeutic drugs. To ensure the anticancer properties, an in vitro assay was performed with "CM-biomimetic peptides" on the non-metastatic colon cancer cell line (HT-29). According to the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay results from peptide candidate treatments at 0-400 µM, the IC50 doses of the CM-biomimetic peptide with no toxic and cancer-cell-penetrating ability, original C. militaris biomimetic peptide (C-ori), against the HT-29 cell line were 114.9 µM at 72 hours. The effects of C-ori compared to the doxorubicin, a conventional chemotherapeutic drug for colon cancer treatment, and the combination effects of both the CM-anticancer peptide and doxorubicin were observed. The results showed that C-ori increased the overall efficiency in the combination treatment with doxorubicin. According to the acridine orange/propidium iodine (AO/PI) staining assay, C-ori can induce apoptosis in HT-29 cells significantly, confirmed by chromatin condensation, membrane blebbing, apoptotic bodies, and late apoptosis which were observed under a fluorescence microscope.
Collapse
|
18
|
adipoSIGHT in Therapeutic Response: Consequences in Osteosarcoma Treatment. Bioengineering (Basel) 2021; 8:bioengineering8060083. [PMID: 34200614 PMCID: PMC8229256 DOI: 10.3390/bioengineering8060083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/02/2022] Open
Abstract
Chemotherapeutic resistance is a major problem in effective cancer treatment. Cancer cells engage various cells or mechanisms to resist anti-cancer therapeutics, which results in metastasis and the recurrence of disease. Considering the cellular heterogeneity of cancer stroma, the involvement of stem cells is reported to affect the proliferation and metastasis of osteosarcoma. Hence, the duo (osteosarcoma: Saos 2 and human adipose-derived stem cells: ASCs) is co-cultured in present study to investigate the therapeutic response using a nonadherent, concave surface. Staining with a cell tracker allows real-time microscopic monitoring of the cell arrangement within the sphere. Cell–cell interaction is investigated by means of E-cadherin expression. Comparatively high expression of E-cadherin and compact organization is observed in heterotypic tumorspheres (Saos 2–ASCs) compared to homotypic ones (ASCs), limiting the infiltration of chemotherapeutic compound doxorubicin into the heterotypic tumorsphere, which in turn protects cells from the toxic effect of the chemotherapeutic. In addition, genes known to be associated with drug resistance, such as SOX2, OCT4, and CD44 are overexpressed in heterotypic tumorspheres post-chemotherapy, indicating that the duo collectively repels the effect of doxorubicin. The interaction between ASCs and Saos 2 in the present study points toward the growing oncological risk of using ASC-based regenerative therapy in cancer patients and warrants further investigation.
Collapse
|
19
|
Cinnamic Acid Derivatives as Cardioprotective Agents against Oxidative and Structural Damage Induced by Doxorubicin. Int J Mol Sci 2021; 22:ijms22126217. [PMID: 34207549 PMCID: PMC8227863 DOI: 10.3390/ijms22126217] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023] Open
Abstract
Doxorubicin (DOX) is a widely used anticancer drug. However, its clinical use is severely limited due to drug-induced cumulative cardiotoxicity, which leads to progressive cardiomyocyte dysfunction and heart failure. Enormous efforts have been made to identify potential strategies to alleviate DOX-induced cardiotoxicity; however, to date, no universal and highly effective therapy has been introduced. Here we reported that cinnamic acid (CA) derivatives exert a multitarget protective effect against DOX-induced cardiotoxicity. The experiments were performed on rat cardiomyocytes (H9c2) and human induced-pluripotent-stem-cell-derived cardiomyocytes (hiPSC-CMs) as a well-established model for cardiac toxicity assessment. CA derivatives protected cardiomyocytes by ameliorating DOX-induced oxidative stress and viability reduction. Our data indicated that they attenuated the chemotherapeutic’s toxicity by downregulating levels of caspase-3 and -7. Pre-incubation of cardiomyocytes with CA derivatives prevented DOX-induced motility inhibition in a wound-healing assay and limited cytoskeleton rearrangement. Detailed safety analyses—including hepatotoxicity, mutagenic potential, and interaction with the hERG channel—were performed for the most promising compounds. We concluded that CA derivatives show a multidirectional protective effect against DOX-induced cardiotoxicity. The results should encourage further research to elucidate the exact molecular mechanism of the compounds’ activity. The lead structure of the analyzed CA derivatives may serve as a starting point for the development of novel therapeutics to support patients undergoing DOX therapy.
Collapse
|
20
|
Synergistic Effect of Doxorubicin and siRNA-Mediated Silencing of Mcl-1 Using Cationic Niosomes against 3D MCF-7 Spheroids. Pharmaceutics 2021; 13:pharmaceutics13040550. [PMID: 33919902 PMCID: PMC8070967 DOI: 10.3390/pharmaceutics13040550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy is a vital option for cancer treatment; however, its therapeutic outcomes are limited by dose-dependent toxicity and the occurrence of chemoresistance. siRNAs have emerged as an attractive therapeutic option enabling specific interference with target genes. Combination therapy using chemotherapeutic agents along with gene therapy could be a potential strategy for cancer management, which not only improves therapeutic efficacy but also decreases untoward effects from dose reduction. In this study, a cationic niosome containing plier-like cationic lipid B was used to convey siRNA against anti-apoptotic mRNA into MCF-7 and MDA-MB-231 cells. Mcl-1 silencing markedly decreased the viability of MCF-7 cells and triggered apoptosis. Moreover, computer modeling suggested that the combination of doxorubicin (Dox) and Mcl-1 siRNA exhibited a synergistic relationship and enabled a dose reduction of each agent at 1.71 and 3.91 folds, respectively, to reach a 90% inhibitory effect when compared to single-agent treatments. Synergistic antitumor activity was further verified in a 3D spheroid culture which revealed, in contrast to single-agent treatment, the combination markedly decreased spheroid volume over time. Together, the combination therapy between Mcl-1 silencing and Dox exhibits a synergistic effect that may be exploited for novel breast cancer treatment.
Collapse
|
21
|
Dao NV, Ercole F, Urquhart MC, Kaminskas LM, Nowell CJ, Davis TP, Sloan EK, Whittaker MR, Quinn JF. Trisulfide linked cholesteryl PEG conjugate attenuates intracellular ROS and collagen-1 production in a breast cancer co-culture model. Biomater Sci 2021; 9:835-846. [PMID: 33231231 DOI: 10.1039/d0bm01544j] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The progression of cancer has been closely-linked with augmentation of cellular reactive oxygen species (ROS) levels and ROS-associated changes in the tumour microenvironment (TME), including alterations to the extracellular matrix and associated low drug uptake. Herein we report the application of a co-culture model to simulate the ROS based cell-cell interactions in the TME using fibroblasts and breast cancer cells, and describe how novel reactive polymers can be used to modulate those interactions. Under the co-culture conditions, both cell types exhibited modifications in behaviour, including significant overproduction of ROS in the cancer cells, and elevation of the collagen-1 secretion and stained actin filament intensity in the fibroblasts. To examine the potential of using reactive antioxidant polymers to intercept ROS communication and thereby manipulate the TME, we employed H2S-releasing macromolecular conjugates which have been previously demonstrated to mitigate ROS production in HEK cells. The specific conjugate used, mPEG-SSS-cholesteryl (T), significantly reduced ROS levels in co-cultured cancer cells by approximately 50%. This reduction was significantly greater than that observed with the other positive antioxidant controls. Exposure to T was also found to downregulate levels of collagen-1 in the co-cultured fibroblasts, while exhibiting less impact on cells in mono-culture. This would suggest a possible downstream effect of ROS-mitigation by T on stromal-tumour cell signalling. Since fibroblast-derived collagens modulate crucial steps in tumorigenesis, this ROS-associated effect could potentially be harnessed to slow cancer progression. The model may also be beneficial for interrogating the impact of antioxidants on naturally enhanced ROS levels, rather than relying on the application of exogenous oxidants to simulate elevated ROS levels.
Collapse
Affiliation(s)
- Nam V Dao
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia. and Department of Physical Chemistry and Physics, Hanoi University of Pharmacy, Hanoi 10000, Vietnam
| | - Francesca Ercole
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - Matthew C Urquhart
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - Lisa M Kaminskas
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Thomas P Davis
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia. and Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Erica K Sloan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia and Peter MacCallum Cancer Centre, Division of Surgery, Melbourne, VIC 3000, Australia
| | - Michael R Whittaker
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia.
| | - John F Quinn
- Australian Research Council - Centre of Excellence in Convergent Bio-Nano Science and Technology, Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia. and Department of Chemical Engineering, Faculty of Engineering, Monash University, Wellington Road, Clayton, Victoria 3800, Australia
| |
Collapse
|
22
|
Sreeja S, Parameshwar R, Varma PRH, Sailaja GS. Hierarchically Porous Osteoinductive Poly(hydroxyethyl methacrylate- co-methyl methacrylate) Scaffold with Sustained Doxorubicin Delivery for Consolidated Osteosarcoma Treatment and Bone Defect Repair. ACS Biomater Sci Eng 2021; 7:701-717. [PMID: 33395260 DOI: 10.1021/acsbiomaterials.0c01628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A bifronted cure system for osteosarcoma, a common aggressive bone tumor, is highly in demand to prevail the postsurgical adversities in connection with systemic chemotherapy and repair of critical-size bone defects. The hierarchically porous therapeutic scaffolds presented here are synthesized by free radical-initiated copolymerization of hydroxyethyl methacrylate and methyl methacrylate [HEMA/MMA 80:20 and 90:10 mM, H2O/NaCl porogen], which are further surface-phosphorylated [P-PHM] and transformed to bifunctional by impregnating doxorubicin (DOX) [DOXP-PHM]. The P-PHM scaffolds exhibited porous microarchitecture analogous to native cancellous bone (scanning electron microscopy analysis), while X-ray photoelectron spectroscopy analysis authenticated surface phosphorylation. Based on pore characteristics, swelling attributes and slow-pace degradation, P-PHM9163 and P-PHM8263 (HEMA/MMA 90:10 and 80:20 with H2O/NaCl: 60/3.0 weight %, respectively) were chosen from the series and evaluated for osteoinductive efficacy in vitro. Both P-PHM9163 and P-PHM8263 invoked calcium phosphate mineralization in simulated physiological conditions (day 14) with Ca/P ratios of 1.58 and 1.66 respectively, comparable to human bone (1.67). Early biomineralization (Alizarin Red S and von Kossa staining) was evidenced at day 7, while osteoblast differentiation was verified by time-dependent expression of the typical late marker, osteocalcin, at day 14 and 21 in rat bone marrow mesenchymal cells. DOX-loaded P-PHM9163 (DOXP-PHM9163) exhibited pH-responsive (tumor analogous pH; 6.5) sustained release of DOX for prolonged time (up to 45 days) and invoked cellular alterations by cortical stress fiber formation and DNA fragmentation in human osteosarcoma cells leading to early apoptosis (24 h), validated by annexin V/PI staining (FACS) and immunostaining (F-actin/DAPI). Subsequent to DOX release tenure, the scaffold induced the formation of well-organized, porous post-release Ca-P apatite coating (Ca/P is 1.3) in simulated body fluid (day 14) which further endorses the dual functionality of the system. Altogether, the results accentuate that DOXP-PHM9163 is a potential bifunctional therapeutic scaffold capable of extended localized chemotherapeutic delivery in-line with inherent osteogenesis for efficient bone cancer treatment.
Collapse
Affiliation(s)
- S Sreeja
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kochi, Kerala 682 022, India
| | - Ramesh Parameshwar
- Division of Polymeric Medical Devices, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695 011, India
| | - P R Harikrishna Varma
- Head of Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram 695 011, India
| | - G S Sailaja
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kochi, Kerala 682 022, India.,Inter University Centre for Nanomaterials and Devices (IUCND), Cochin University of Science and Technology, Kochi, Kerala 682 022, India.,Centre for Excellence in Advanced Materials, Cochin University of Science and Technology, Kochi, Kerala 682 022, India
| |
Collapse
|
23
|
Zarate SM, Pandey G, Chilukuri S, Garcia JA, Cude B, Storey S, Salem NA, Bancroft EA, Hook M, Srinivasan R. Cytisine is neuroprotective in female but not male 6-hydroxydopamine lesioned parkinsonian mice and acts in combination with 17-β-estradiol to inhibit apoptotic endoplasmic reticulum stress in dopaminergic neurons. J Neurochem 2021; 157:710-726. [PMID: 33354763 DOI: 10.1111/jnc.15282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/11/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022]
Abstract
Apoptotic endoplasmic reticulum (ER) stress is a major mechanism for dopaminergic (DA) loss in Parkinson's disease (PD). We assessed if low doses of the partial α4β2 nicotinic acetylcholine receptor agonist, cytisine attenuates apoptotic ER stress and exerts neuroprotection in substantia nigra pars compacta (SNc) DA neurons. Alternate day intraperitoneal injections of 0.2 mg/kg cytisine were administered to female and male mice with 6-hydroxydopamine (6-OHDA) lesions in the dorsolateral striatum, which caused unilateral degeneration of SNc DA neurons. Cytisine attenuated 6-OHDA-induced PD-related behaviors in female, but not in male mice. We also found significant reductions in tyrosine hydroxylase (TH) loss within the lesioned SNc of female, but not male mice. In contrast to female mice, DA neurons within the lesioned SNc of male mice showed a cytisine-induced pathological increase in the nuclear translocation of the pro-apoptotic ER stress protein, C/EBP homologous protein (CHOP). To assess the role of estrogen in cytisine neuroprotection in female mice, we exposed primary mouse DA cultures to either 10 nM 17-β-estradiol and 200 nM cytisine or 10 nM 17-β-estradiol alone. 17-β-estradiol reduced expression of CHOP, whereas cytisine exposure reduced 6-OHDA-mediated nuclear translocation of two other ER stress proteins, activating transcription factor 6 and x-box-binding protein 1, but not CHOP. Taken together, these data show that cytisine and 17-β-estradiol work in combination to inhibit all three arms (activating transcription factor 6, x-box-binding protein 1, and CHOP) of apoptotic ER stress signaling in DA neurons, which can explain the neuroprotective effect of low-dose cytisine in female mice.
Collapse
Affiliation(s)
- Sara M Zarate
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Gauri Pandey
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Sunanda Chilukuri
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Jose A Garcia
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Brittany Cude
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Shannon Storey
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Nihal A Salem
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA.,Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA
| | - Eric A Bancroft
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA
| | - Michelle Hook
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA.,Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, College of Medicine, Texas A&M University, Bryan, TX, USA.,Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA
| |
Collapse
|
24
|
Pop RM, Bocsan IC, Buzoianu AD, Chedea VS, Socaci SA, Pecoraro M, Popolo A. Evaluation of the Antioxidant Activity of Nigella sativa L. and Allium ursinum Extracts in a Cellular Model of Doxorubicin-Induced Cardiotoxicity. Molecules 2020; 25:molecules25225259. [PMID: 33187371 PMCID: PMC7697550 DOI: 10.3390/molecules25225259] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/07/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Natural products black cumin—Nigella sativa (N. sativa) and wild garlic—Allium ursinum (AU) are known for their potential role in reducing cardiovascular risk factors, including antracycline chemotherapy. Therefore, this study investigates the effect of N. sativa and AU water and methanolic extracts in a cellular model of doxorubicin (doxo)-induced cardiotoxicity. The extracts were characterized using Ultraviolet-visible (UV-VIS) spectroscopy, Fourier-transform infrared (FT-IR) spectroscopy, Liquid Chromatography coupled with Mass Spectrometry (LC-MS) and Gas Chromatography coupled with Mass Spectrometry (GC-MS) techniques. Antioxidant activity was evaluated on H9c2 cells. Cytosolic and mitochondrial reactive oxygen species (ROS) release was evaluated using 2′,7′-dichlorofluorescin-diacetate (DHCF-DA) and mitochondria-targeted superoxide indicator (MitoSOX red), respectively. Mitochondrial membrane depolarization was evaluated by flow cytometry. LC-MS analysis identified 12 and 10 phenolic compounds in NSS and AU extracts, respectively, with flavonols as predominant compounds. FT-IR analysis identified the presence of carbohydrates, amino acids and lipids in both plants. GC-MS identified the sulfur compounds in the AU water extract. N. sativa seeds (NSS) methanolic extract had the highest antioxidant activity reducing both intracellular and mitochondrial ROS release. All extracts (excepting AU methanolic extract) preserved H9c2 cells viability. None of the investigated plants affected the mitochondrial membrane depolarization. N. sativa and AU are important sources of bioactive compounds with increased antioxidant activities, requiring different extraction solvents to obtain the pharmacological effects.
Collapse
Affiliation(s)
- Raluca Maria Pop
- Department of Pharmacology, Toxicology and Clinical Pharmacology, “Iuliu Hatieganu” University of Medicine and Pharmacy, Victor Babes, No 8, 400012 Cluj-Napoca, Romania; (I.C.B.); (A.D.B.)
- Correspondence: (R.M.P.); (A.P.)
| | - Ioana Corina Bocsan
- Department of Pharmacology, Toxicology and Clinical Pharmacology, “Iuliu Hatieganu” University of Medicine and Pharmacy, Victor Babes, No 8, 400012 Cluj-Napoca, Romania; (I.C.B.); (A.D.B.)
| | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, “Iuliu Hatieganu” University of Medicine and Pharmacy, Victor Babes, No 8, 400012 Cluj-Napoca, Romania; (I.C.B.); (A.D.B.)
| | - Veronica Sanda Chedea
- Research Station for Viticulture and Enology Blaj (SCDVV Blaj), 515400 Blaj, Romania;
| | - Sonia Ancuța Socaci
- Department of Food Science, University of Agricultural Sciences and Veterinary Medicine of Cluj-Napoca, Calea Manaștur 3–5, 400372 Cluj-Napoca, Romania;
| | - Michela Pecoraro
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy;
| | - Ada Popolo
- Department of Pharmacy, University of Salerno, via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy;
- Correspondence: (R.M.P.); (A.P.)
| |
Collapse
|
25
|
Popescu RC, Straticiuc M, Mustăciosu C, Temelie M, Trușcă R, Vasile BȘ, Boldeiu A, Mirea D, Andrei RF, Cenușă C, Mogoantă L, Mogoșanu GD, Andronescu E, Radu M, Veldwijk MR, Savu DI. Enhanced Internalization of Nanoparticles Following Ionizing Radiation Leads to Mitotic Catastrophe in MG-63 Human Osteosarcoma Cells. Int J Mol Sci 2020; 21:ijms21197220. [PMID: 33007844 PMCID: PMC7583846 DOI: 10.3390/ijms21197220] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/27/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
This study aims to investigate whether ionizing radiation combined with doxorubicin-conjugated iron oxide nanoparticles (NP-DOX) improves the internalization and cytotoxic effects of the nano-carrier-mediated drug delivery in MG-63 human osteosarcoma cells. NP-DOX was designed and synthesized using the co-precipitation method. Highly stable and crystalline nanoparticles conjugated with DOX were internalized in MG-63 cells through macropinocytosis and located in the perinuclear area. Higher nanoparticles internalization in MG-63 cells previously exposed to 1 Gy X-rays was correlated with an early accumulation of cells in G2/M, starting at 12 h after treatment. After 48 h, the application of the combined treatment led to higher cytotoxic effects compared to the individual treatment, with a reduction in the metabolic capacity and unrepaired DNA breaks, whilst a low percent of arrested cells, contributing to the commitment of mitotic catastrophe. NP-DOX showed hemocompatibility and no systemic cytotoxicity, nor histopathological alteration of the main organs.
Collapse
Affiliation(s)
- Roxana Cristina Popescu
- Department of Life and Environmental Physics, “Horia Hulubei” National Insitute of Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Magurele, Romania; (R.C.P.); (C.M.); (M.T.); (M.R.)
- Department of Science and Engineering of Oxide Materials and Nanomaterials, “Politehnica” University of Bucharest (UPB), 1-7 Polizu Street, 011061 Bucharest, Romania;
| | - Mihai Straticiuc
- Department of Applied Nuclear Physics, “Horia Hulubei” National Insitute of Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.M.); (R.F.A.)
| | - Cosmin Mustăciosu
- Department of Life and Environmental Physics, “Horia Hulubei” National Insitute of Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Magurele, Romania; (R.C.P.); (C.M.); (M.T.); (M.R.)
- Department of Science and Engineering of Oxide Materials and Nanomaterials, “Politehnica” University of Bucharest (UPB), 1-7 Polizu Street, 011061 Bucharest, Romania;
| | - Mihaela Temelie
- Department of Life and Environmental Physics, “Horia Hulubei” National Insitute of Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Magurele, Romania; (R.C.P.); (C.M.); (M.T.); (M.R.)
| | - Roxana Trușcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National Research Center for Micro and Nanomaterials, “Politehnica” University of Bucharest (UPB), 313 Splaiul Independenţei, 060042 Bucharest, Romania; (R.T.); (B.Ș.V.)
| | - Bogdan Ștefan Vasile
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National Research Center for Micro and Nanomaterials, “Politehnica” University of Bucharest (UPB), 313 Splaiul Independenţei, 060042 Bucharest, Romania; (R.T.); (B.Ș.V.)
| | - Adina Boldeiu
- Laboratory of Nanobiotechnology, National Institute for Research and Development in Microtechnologies (IMT), 12A Erou Iancu Nicolae Street, 077190 Bucharest, Romania;
| | - Dragoş Mirea
- Department of Applied Nuclear Physics, “Horia Hulubei” National Insitute of Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.M.); (R.F.A.)
| | - Radu Florin Andrei
- Department of Applied Nuclear Physics, “Horia Hulubei” National Insitute of Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Magurele, Romania; (M.S.); (D.M.); (R.F.A.)
- Department of Physics, Applied Science Faculty, “Politehnica” University of Bucharest (UPB), 303 Splaiul Independentei, 060042 Bucharest, Romania
| | - Constantin Cenușă
- Radioisotopes and Radiation Metrology Department, “Horia Hulubei” National Insitute of Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Magurele, Romania;
| | - Laurenţiu Mogoantă
- Research Center for Microscopic Morphology and Immunology, University of Medicine and Pharmacy of Craiova (UMFCV), 2 Petru Rareș Street, 200349 Craiova, Romania;
| | - George Dan Mogoșanu
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova (UMFCV), 2 Petru Rareș Street, 200349 Craiova, Romania;
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, “Politehnica” University of Bucharest (UPB), 1-7 Polizu Street, 011061 Bucharest, Romania;
| | - Mihai Radu
- Department of Life and Environmental Physics, “Horia Hulubei” National Insitute of Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Magurele, Romania; (R.C.P.); (C.M.); (M.T.); (M.R.)
| | - Marlon R. Veldwijk
- Department of Radiation Oncology, Universitätsmedizin Mannheim (UMM), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
- Correspondence: (M.R.V.); (D.I.S.); Tel.: +49-621-383-3750 (M.R.V.); +40-214-046-134 (D.I.S.)
| | - Diana Iulia Savu
- Department of Life and Environmental Physics, “Horia Hulubei” National Insitute of Physics and Nuclear Engineering (IFIN-HH), 30 Reactorului Street, 077125 Magurele, Romania; (R.C.P.); (C.M.); (M.T.); (M.R.)
- Correspondence: (M.R.V.); (D.I.S.); Tel.: +49-621-383-3750 (M.R.V.); +40-214-046-134 (D.I.S.)
| |
Collapse
|
26
|
Oltolina F, Peigneux A, Colangelo D, Clemente N, D’Urso A, Valente G, Iglesias GR, Jiménez-Lopez C, Prat M. Biomimetic Magnetite Nanoparticles as Targeted Drug Nanocarriers and Mediators of Hyperthermia in an Experimental Cancer Model. Cancers (Basel) 2020; 12:cancers12092564. [PMID: 32916816 PMCID: PMC7564965 DOI: 10.3390/cancers12092564] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/03/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The application of simultaneous and different strategies to treat cancer appears a promising therapeutic approach. Herein we proposed the application of chemotherapy combined with a magnetic nanocarrier delivery system to an in vitro and an in vivo experimental mammary carcinoma model. Drug-loaded biomimetic magnetic nanoparticle can be directed and concentrated on the tumor cells or site by the apposition of a magnet. Moreover, these nanoparticles can respond to an alternating magnetic field by developing hyperthermia around 43 °C, a temperature at which tumor cells, but not healthy cells, are particularly sensitive and thus induced to death. Indeed, when this nanoformulation is injected in vivo in the tumor site, and hyperthermia is generated, the combined chemo-thermal therapy mediated by these drug-loaded magnetic nanoparticles have a stronger therapeutic benefit compared to that carried out by the chemotherapeutic alone. These nanoformulation and strategy are thus promising tools for translational applications in cancer therapy. Abstract Biomimetic magnetic nanoparticles mediated by magnetosome proteins (BMNPs) are potential innovative tools for cancer therapy since, besides being multifunctional platforms, they can be manipulated by an external gradient magnetic field (GMF) and/or an alternating magnetic field (AMF), mediating targeting and hyperthermia, respectively. We evaluated the cytocompatibility/cytotoxicity of BMNPs and Doxorubicin (DOXO)-BMNPs in the presence/absence of GMF in 4T1 and MCF-7 cells as well as their cellular uptake. We analyzed the biocompatibility and in vivo distribution of BMNPs as well as the effect of DOXO-BMNPs in BALB/c mice bearing 4T1 induced mammary carcinomas after applying GMF and AMF. Results: GMF enhanced the cell uptake of both BMNPs and DOXO-BMNPs and the cytotoxicity of DOXO-BMNPs. BMNPs were biocompatible when injected intravenously in BALB/c mice. The application of GMF on 4T1 tumors after each of the repeated (6×) iv administrations of DOXO-BMNPs enhanced tumor growth inhibition when compared to any other treatment, including that with soluble DOXO. Moreover, injection of DOXO-BMNPs in the tumor combined with application of an AMF resulted in a significant tumor weight reduction. These promising results show the suitability of BMNPs as magnetic nanocarriers for local targeted chemotherapy and as local agents for hyperthermia.
Collapse
Affiliation(s)
- Francesca Oltolina
- Department of Health Sciences, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy; (F.O.); (D.C.); (N.C.); (A.D.)
| | - Ana Peigneux
- Department of Microbiology, University of Granada, Campus Fuentenueva, s/n, 18071 Granada, Spain;
| | - Donato Colangelo
- Department of Health Sciences, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy; (F.O.); (D.C.); (N.C.); (A.D.)
| | - Nausicaa Clemente
- Department of Health Sciences, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy; (F.O.); (D.C.); (N.C.); (A.D.)
| | - Annarita D’Urso
- Department of Health Sciences, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy; (F.O.); (D.C.); (N.C.); (A.D.)
| | - Guido Valente
- Department of Translational Medicine, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy;
| | - Guillermo R. Iglesias
- Department of Applied Physic, University of Granada, Campus Fuentenueva, s/n, 18071 Granada, Spain;
| | - Concepcion Jiménez-Lopez
- Department of Microbiology, University of Granada, Campus Fuentenueva, s/n, 18071 Granada, Spain;
- Correspondence: (C.J.-L.); (M.P.)
| | - Maria Prat
- Department of Health Sciences, Università del Piemonte Orientale A. Avogadro, Via Solaroli 17, 28100 Novara, Italy; (F.O.); (D.C.); (N.C.); (A.D.)
- Centro di Biotecnologie per la Ricerca Medica Applicata (BRMA), Via Solaroli 17, 28100 Novara, Italy
- Consorzio Interuniversitario per Biotecnologie (CIB), Località Padriciano 99, 34149 Area di Ricerca, Trieste, Italy
- Consorzio Interuniversitario Nazionale per la Scienza e Tecnologia dei Materiali (INSTM), Via Giuseppe Giusti 9, 50121 Firenze, Italy
- Consorzio Interuniversitario di Ricerca in Chimica dei Metalli nei Sistemi Biologici (CIRCMSB) Piazza Umberto I 1, 70121 Bari, Italy
- Centro Interdipartimentale di Medicina Rigenerativa (CIMeR), Via Montpellier, 1, 00133 Roma, Italy
- Correspondence: (C.J.-L.); (M.P.)
| |
Collapse
|
27
|
Norouzi M, Yathindranath V, Thliveris JA, Kopec BM, Siahaan TJ, Miller DW. Doxorubicin-loaded iron oxide nanoparticles for glioblastoma therapy: a combinational approach for enhanced delivery of nanoparticles. Sci Rep 2020; 10:11292. [PMID: 32647151 PMCID: PMC7347880 DOI: 10.1038/s41598-020-68017-y] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 03/16/2020] [Indexed: 01/05/2023] Open
Abstract
Although doxorubicin (DOX) is an effective anti-cancer drug with cytotoxicity in a variety of different tumors, its effectiveness in treating glioblastoma multiforme (GBM) is constrained by insufficient penetration across the blood–brain barrier (BBB). In this study, biocompatible magnetic iron oxide nanoparticles (IONPs) stabilized with trimethoxysilylpropyl-ethylenediamine triacetic acid (EDT) were developed as a carrier of DOX for GBM chemotherapy. The DOX-loaded EDT-IONPs (DOX-EDT-IONPs) released DOX within 4 days with the capability of an accelerated release in acidic microenvironments. The DOX-loaded EDT-IONPs (DOX-EDT-IONPs) demonstrated an efficient uptake in mouse brain-derived microvessel endothelial, bEnd.3, Madin–Darby canine kidney transfected with multi-drug resistant protein 1 (MDCK-MDR1), and human U251 GBM cells. The DOX-EDT-IONPs could augment DOX’s uptake in U251 cells by 2.8-fold and significantly inhibited U251 cell proliferation. Moreover, the DOX-EDT-IONPs were found to be effective in apoptotic-induced GBM cell death (over 90%) within 48 h of treatment. Gene expression studies revealed a significant downregulation of TOP II and Ku70, crucial enzymes for DNA repair and replication, as well as MiR-155 oncogene, concomitant with an upregulation of caspase 3 and tumor suppressors i.e., p53, MEG3 and GAS5, in U251 cells upon treatment with DOX-EDT-IONPs. An in vitro MDCK-MDR1-GBM co-culture model was used to assess the BBB permeability and anti-tumor activity of the DOX-EDT-IONPs and DOX treatments. While DOX-EDT-IONP showed improved permeability of DOX across MDCK-MDR1 monolayers compared to DOX alone, cytotoxicity in U251 cells was similar in both treatment groups. Using a cadherin binding peptide (ADTC5) to transiently open tight junctions, in combination with an external magnetic field, significantly enhanced both DOX-EDT-IONP permeability and cytotoxicity in the MDCK-MDR1-GBM co-culture model. Therefore, the combination of magnetic enhanced convective diffusion and the cadherin binding peptide for transiently opening the BBB tight junctions are expected to enhance the efficacy of GBM chemotherapy using the DOX-EDT-IONPs. In general, the developed approach enables the chemotherapeutic to overcome both BBB and multidrug resistance (MDR) glioma cells while providing site-specific magnetic targeting.
Collapse
Affiliation(s)
- Mohammad Norouzi
- Department of Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada.,Department of Pharmacology and Therapeutics, University of Manitoba, A205 Chown Bldg., 753 McDermot Avenue, Winnipeg, MB, Canada
| | - Vinith Yathindranath
- Department of Pharmacology and Therapeutics, University of Manitoba, A205 Chown Bldg., 753 McDermot Avenue, Winnipeg, MB, Canada
| | - James A Thliveris
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada
| | - Brian M Kopec
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Teruna J Siahaan
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, USA
| | - Donald W Miller
- Department of Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada. .,Department of Pharmacology and Therapeutics, University of Manitoba, A205 Chown Bldg., 753 McDermot Avenue, Winnipeg, MB, Canada.
| |
Collapse
|
28
|
Newell M, Patel D, Goruk S, Field CJ. Docosahexaenoic Acid Incorporation Is Not Affected by Doxorubicin Chemotherapy in either Whole Cell or Lipid Raft Phospholipids of Breast Cancer Cells in vitro and Tumor Phospholipids in vivo. Lipids 2020; 55:549-565. [PMID: 32588470 DOI: 10.1002/lipd.12252] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 11/07/2022]
Abstract
To better understand how docosahexaenoic acid (DHA) improves the effects of doxorubicin (DOX), we examined DHA ± DOX on changes in whole cell and lipid raft phospholipids (PL) of MDA-MB-231 and MCF-7 breast cancer cells. We sought to confirm whether the relative changes in PL DHA content of MDA-MB-231 cells could be extended to PL from MDA-MB-231 tumors grown in mice fed a DHA supplemented diet ±DOX. Treatment with DHA did not change PL composition yet DOX increased the proportion of phosphatidylserine in MCF-7 cell lipid rafts by two-fold (p < 0.001). Regardless of DOX, the relative percent incorporation of DHA was higher in MDA-MB-231 cells compared to MCF-7 cells in phosphatidylserine, phosphatidylethanolamine, and phosphatidylcholine (whole cell and lipid rafts); and higher in phosphatidylethanolamine vs. phosphatidylcholine (4.4-fold in MCF-7 and 6-fold in MDA-MB-231 cells respectively). DHA treatment increased eicosapentaenoic acid and docosapentaenoic acid in MDA-MB-231 cells but not MCF-7 cells. Increased DHA content in MDA-MB-231 cells, MCF-7 cells, and MDA-MB-231 tumors in all PL moieties (except sphingomyelin) corresponded with reduced arachidonic acid (p < 0.05). Feeding mice 2.8% (w/w of fat) DHA ± DOX increased tumor necrotic regions (p < 0.05). This study established differential incorporation of DHA into whole cell and lipid rafts between human breast cancer cell lines. However, within each cell line, this incorporation was not altered by DOX confirming that DOX does not change membrane lipid composition. Furthermore, our findings indicate that membrane changes observed in vitro are translatable to in vivo changes and that DHA + DOX could contribute to the anticancer effects through increased necrosis.
Collapse
Affiliation(s)
- Marnie Newell
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Dhruvesh Patel
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Susan Goruk
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Catherine J Field
- Department of Agricultural, Food and Nutritional Science, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| |
Collapse
|
29
|
Zhu J, Singh M, Selivanova G, Peuget S. Pifithrin-α alters p53 post-translational modifications pattern and differentially inhibits p53 target genes. Sci Rep 2020; 10:1049. [PMID: 31974452 PMCID: PMC6978515 DOI: 10.1038/s41598-020-58051-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/06/2020] [Indexed: 11/19/2022] Open
Abstract
Pifithrin-α (PFT-α) is a small molecule which has been widely used as a specific inhibitor of p53 transcription activity. However, its molecular mechanism of action remains unclear. PFT-α has also been described to display potent p53-independent activity in cells. In this study, we addressed the mechanism of action of PFT-α. We found that PFT-α failed to prevent the effects of Mdm2 inhibitor Nutlin-3 on cell cycle and apoptosis in several cancer cell lines. However, PFT-α rescued normal primary fibroblasts from growth inhibition by Nutlin-3. PFT-α displayed a very limited effect on p53-dependent transcription upon its activation by Nutlin-3. Moreover, PFT-α inhibitory effect on transcription was highly dependent on the nature of the p53 target gene. PFT-α attenuated post-translational modifications of p53 without affecting total p53 protein level. Finally, we found that PFT-α can decrease the level of intracellular reactive oxygen species through activation of an aryl hydrocarbon receptor (AHR)-Nrf2 axis in a p53-independent manner. In conclusion, PFT-α inhibits only some aspects of p53 function, therefore it should be used with extreme caution to study p53-dependent processes.
Collapse
Affiliation(s)
- Jiawei Zhu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Madhurendra Singh
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Galina Selivanova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Sylvain Peuget
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
30
|
Santos SM, Hartman JL. A yeast phenomic model for the influence of Warburg metabolism on genetic buffering of doxorubicin. Cancer Metab 2019; 7:9. [PMID: 31660150 PMCID: PMC6806529 DOI: 10.1186/s40170-019-0201-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 09/03/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The influence of the Warburg phenomenon on chemotherapy response is unknown. Saccharomyces cerevisiae mimics the Warburg effect, repressing respiration in the presence of adequate glucose. Yeast phenomic experiments were conducted to assess potential influences of Warburg metabolism on gene-drug interaction underlying the cellular response to doxorubicin. Homologous genes from yeast phenomic and cancer pharmacogenomics data were analyzed to infer evolutionary conservation of gene-drug interaction and predict therapeutic relevance. METHODS Cell proliferation phenotypes (CPPs) of the yeast gene knockout/knockdown library were measured by quantitative high-throughput cell array phenotyping (Q-HTCP), treating with escalating doxorubicin concentrations under conditions of respiratory or glycolytic metabolism. Doxorubicin-gene interaction was quantified by departure of CPPs observed for the doxorubicin-treated mutant strain from that expected based on an interaction model. Recursive expectation-maximization clustering (REMc) and Gene Ontology (GO)-based analyses of interactions identified functional biological modules that differentially buffer or promote doxorubicin cytotoxicity with respect to Warburg metabolism. Yeast phenomic and cancer pharmacogenomics data were integrated to predict differential gene expression causally influencing doxorubicin anti-tumor efficacy. RESULTS Yeast compromised for genes functioning in chromatin organization, and several other cellular processes are more resistant to doxorubicin under glycolytic conditions. Thus, the Warburg transition appears to alleviate requirements for cellular functions that buffer doxorubicin cytotoxicity in a respiratory context. We analyzed human homologs of yeast genes exhibiting gene-doxorubicin interaction in cancer pharmacogenomics data to predict causality for differential gene expression associated with doxorubicin cytotoxicity in cancer cells. This analysis suggested conserved cellular responses to doxorubicin due to influences of homologous recombination, sphingolipid homeostasis, telomere tethering at nuclear periphery, actin cortical patch localization, and other gene functions. CONCLUSIONS Warburg status alters the genetic network required for yeast to buffer doxorubicin toxicity. Integration of yeast phenomic and cancer pharmacogenomics data suggests evolutionary conservation of gene-drug interaction networks and provides a new experimental approach to model their influence on chemotherapy response. Thus, yeast phenomic models could aid the development of precision oncology algorithms to predict efficacious cytotoxic drugs for cancer, based on genetic and metabolic profiles of individual tumors.
Collapse
Affiliation(s)
- Sean M. Santos
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL USA
| | - John L. Hartman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
31
|
Wang J, Yao Y, Liu X, Wang K, Zhou Q, Tang Y. Protective effects of lycium barbarum polysaccharides on blood-retinal barrier via ROCK1 pathway in diabetic rats. Am J Transl Res 2019; 11:6304-6315. [PMID: 31737184 PMCID: PMC6834516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/27/2019] [Indexed: 06/10/2023]
Abstract
Lycium barbarum polysaccharides (LBP) is commonly known as a traditional Chinese medicine, which has protective effects against diabetic complications in clinic, such as diabetic retinopathy (DR). Previous studies have revealed that Rho/ROCK pathway play an important role in DR development. However, the mechanism between LBP and DR remains unknown. This study aims to explore the clear mechanism of the protective effect of LBP in diabetic retinopathy. In this study, streptozocin (STZ, 45 mg/kg) was administered for diabetic rats modeling. Weight, blood glucose levels and blood lipid were measured to assess the metabolic changes by LBP on diabetic rats. Evans blue (EB) extravasation was determined to assess blood-retinal barrier (BRB) disruption. Hematoxylin and Eosin (HE) staining and immunohistochemistry assay were applied for retina morphology exploration. The membranous disks of retina were examined by transmission electron microscope. Further, high glucose condition was induced in choroidal-retinal endothelial cells (RF/6A). Western blotting was performed for P-Occludin, ROCK1 and P-MLC protein expression. The results indicated that the blood glucose levels, blood lipid and EB infiltration capacity were decreased while the weight was increased in LBP-treated diabetic rats compared with model rats. Moreover, LBP could thicken the overall retina, prevent the disturbance of photoreceptor cell membranous disks and inhibit pathological angiogenesis in diabetes. In addition, the decreased expression of P-Occludin and increased expression of RhoA-associated protein kinase (ROCK) or phosphorylated myosin light chain (P-MLC) were observed in retinal tissue of diabetic rats and high glucose induced by RF/6A cells, which could be rescued by LBP and/or Fasudil. LBP has the protective effects on blood-retinal barrier by regulating the Rho/ROCK signaling pathway in diabetic rats. LBP may be served as a Rho/ROCK inhibitor for the treatment of DR.
Collapse
Affiliation(s)
- Jihong Wang
- Department of Ophthalmology, Affiliated Hospital of Jiangnan University, Wuxi Hospital of Traditional Chinese MedicineWuxi 214000, Jiangsu, China
| | - Yong Yao
- Department of Ophthalmology, Affiliated Wuxi People’s Hospital of Nanjing Medical UniversityWuxi 214000, Jiangsu, China
| | - Xuezheng Liu
- Department of Anatomy, Jinzhou Medical UniversityJinzhou 121001, Liaoning, China
| | - Kelei Wang
- Department of Ophthalmology, Wuxi Hospital of Traditional Chinese MedicineWuxi 214000, Jiangsu, China
| | - Qianqian Zhou
- Department of Ophthalmology, Wuxi Hospital of Traditional Chinese MedicineWuxi 214000, Jiangsu, China
| | - Ying Tang
- Department of Ophthalmology, Wuxi Hospital of Traditional Chinese MedicineWuxi 214000, Jiangsu, China
| |
Collapse
|
32
|
Liver Cancer Cell Lines Treated with Doxorubicin under Normoxia and Hypoxia: Cell Viability and Oncologic Protein Profile. Cancers (Basel) 2019; 11:cancers11071024. [PMID: 31330834 PMCID: PMC6678640 DOI: 10.3390/cancers11071024] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/09/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma is often treated with a combination of doxorubicin and embolization, exposing it to high concentrations and hypoxia. Separation of the possible synergistic effect of this combination in vivo is difficult. Here, treatment with doxorubicin, under hypoxia or normoxia in different liver cancer cell lines, was evaluated. Liver cancer cells HepG2, Huh7, and SNU449 were exposed to doxorubicin, hypoxia, or doxorubicin + hypoxia with different duration. Treatment response was evaluated with cell viability, apoptosis, oxidative stress, and summarized with IC50. The protein profile of a 92-biomarker panel was analyzed on cells treated with 0 or 0.1 µM doxorubicin during 6 or 72 h, under normoxia or hypoxia. Hypoxia decreased viability of HepG2 and SNU499. HepG2 was least and SNU449 most tolerant to doxorubicin treatment. Cytotoxicity of doxorubicin increased over time in HepG2 and Huh7. The combination of doxorubicin + hypoxia affected the cells differently. Normalized protein expression was lower for HepG2 than Huh7 and SNU449. Hierarchical clustering separated HepG2 from Huh7 and SNU449. These three commonly used cell lines have critically different responses to chemotherapy and hypoxia, which was reflected in their different protein expression profile. These different responses suggest that tumors can respond differently to the combination of local chemotherapy and embolization.
Collapse
|
33
|
Zahiri M, Babaei M, Abnous K, Taghdisi SM, Ramezani M, Alibolandi M. Hybrid nanoreservoirs based on dextran‐capped dendritic mesoporous silica nanoparticles for CD133‐targeted drug delivery. J Cell Physiol 2019; 235:1036-1050. [DOI: 10.1002/jcp.29019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Mahsa Zahiri
- Student Research Committee Mashhad University of Medical Sciences Mashhad Iran
- Department of Pharmaceutical Nanotechnology, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Babaei
- Pharmaceutical Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
| | - Khalil Abnous
- Pharmaceutical Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
- Department of Medicinal Chemistry, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute Mashhad University of Medical Sciences Mashhad Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
34
|
Alhowail AH, Bloemer J, Majrashi M, Pinky PD, Bhattacharya S, Yongli Z, Bhattacharya D, Eggert M, Woodie L, Buabeid MA, Johnson N, Broadwater A, Smith B, Dhanasekaran M, Arnold RD, Suppiramaniam V. Doxorubicin-induced neurotoxicity is associated with acute alterations in synaptic plasticity, apoptosis, and lipid peroxidation. Toxicol Mech Methods 2019; 29:457-466. [PMID: 31010378 DOI: 10.1080/15376516.2019.1600086] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cognitive deficits are commonly reported by patients following treatment with chemotherapeutic agents. Anthracycline-containing chemotherapy regimens are associated with cognitive impairment and reductions in neuronal connectivity in cancer survivors, and doxorubicin (Dox) is a commonly used anthracycline. Although it has been reported that Dox distribution to the central nervous system (CNS) is limited, considerable Dox concentrations are observed in the brain with co-administration of certain medications. Additionally, pro-inflammatory cytokines, which are overproduced in cancer or in response to chemotherapy, can reduce the integrity of the blood-brain barrier (BBB). Therefore, the aim of this study was to evaluate the acute neurotoxic effects of Dox on hippocampal neurons. In this study, we utilized a hippocampal cell line (H19-7/IGF-IR) along with rodent hippocampal slices to evaluate the acute neurotoxic effects of Dox. Hippocampal slices were used to measure long-term potentiation (LTP), and expression of proteins was determined by immunoblotting. Cellular assays for mitochondrial complex activity and lipid peroxidation were also utilized. We observed reduction in LTP in hippocampal slices with Dox. In addition, lipid peroxidation was increased as measured by thiobarbituric acid reactive substances content indicating oxidative stress. Caspase-3 expression was increased indicating an increased propensity for cell death. Finally, the phosphorylation of signaling molecules which modulate LTP including extracellular signal-regulated kinase 1/2 (ERK1/2), p38 mitogen-activated protein kinase, and Akt were increased. This data indicates that acute Dox exposure dose-dependently impairs synaptic processes associated with hippocampal neurotransmission, induces apoptosis, and increases lipid peroxidation leading to neurotoxicity.
Collapse
Affiliation(s)
- Ahmad H Alhowail
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Jenna Bloemer
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Mohammed Majrashi
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Priyanka D Pinky
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | | | - Zhang Yongli
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA.,b Tianjin Huanhu Hospital , Tianjin , PR China
| | - Dwipayan Bhattacharya
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Matthew Eggert
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Lauren Woodie
- c Department of Nutrition, Dietetics and Hospitality Management , College of Human Sciences, Auburn University , Auburn , AL , USA
| | - Manal A Buabeid
- d College of Pharmacy and Health Sciences , Ajman University , Ajman , UAE
| | - Nathaniel Johnson
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Alyssa Broadwater
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Bruce Smith
- e Department of Anatomy, Physiology and Pharmacology , College of Veterinary Medicine, Auburn University , Auburn , AL , USA
| | | | - Robert D Arnold
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| | - Vishnu Suppiramaniam
- a Department of Drug Discovery and Development , Auburn University , Auburn , AL , USA
| |
Collapse
|
35
|
Hamidu A, Mokrish A, Mansor R, Razak ISA, Danmaigoro A, Jaji AZ, Bakar ZA. Modified methods of nanoparticles synthesis in pH-sensitive nano-carriers production for doxorubicin delivery on MCF-7 breast cancer cell line. Int J Nanomedicine 2019; 14:3615-3627. [PMID: 31190815 PMCID: PMC6535674 DOI: 10.2147/ijn.s190830] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 02/22/2019] [Indexed: 12/16/2022] Open
Abstract
Purpose: Modified top-down procedure was successfully employed in the synthesis of aragonite nanoparticles (NPs) from cheaply available natural seawater cockle shells. This was with the aim of developing a pH-sensitive nano-carrier for effective delivery of doxorubicin (DOX) on MCF-7 breast cancer cell line. Methods: The shells were cleaned with banana pelts, ground using a mortar and pestle, and stirred vigorously on a rotary pulverizing blending machine in dodecyl dimethyl betane solution. This simple procedure avoids the use of stringent temperatures and unsafe chemicals associated with NP production. The synthesized NPs were loaded with DOX to form DOX-NPs. The free and DOX-loaded NPs were characterized for physicochemical properties using field emission scanning electron microscopy, transmission electron microscopy, zeta potential analysis, Fourier transform infrared spectroscopy, and X-ray diffraction. The release profile, cytotoxicity, and cell uptake were evaluated. Results: NPs had an average diameter of 35.50 nm, 19.3% loading content, 97% encapsulation efficiency, and a surface potential and intensity of 19.1±3.9 mV and 100%, respectively. A slow and sustained pH-specific controlled discharge profile of DOX from DOX-NPs was observed, clearly showing apoptosis/necrosis induced by DOX-NPs through endocytosis. The DOX-NPs had IC50 values 1.829, 0.902, and 1.0377 µg/mL at 24, 48, and 72 hrs, while those of DOX alone were 0.475, 0.2483, and 0.0723 µg/mL, respectively. However, even at higher concentration, no apparent toxicity was observed with the NPs, revealing their compatibility with MCF-7 cells with a viability of 92%. Conclusions: The modified method of NPs synthesis suggests the tremendous potential of the NPs as pH-sensitive nano-carriers in cancer management because of their pH targeting ability toward cancerous cells.
Collapse
Affiliation(s)
- Ahmed Hamidu
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.,Department of Sciences and Engineering, Federal Polytechnic Mubi, Adamawa State, Nigeria
| | - Ajat Mokrish
- Department of Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor 434000, Selangor, Malaysia
| | - Rozaihan Mansor
- Department of Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor 434000, Selangor, Malaysia
| | - Intan Shameha Abdul Razak
- Department of Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor 434000, Selangor, Malaysia
| | - Abubakar Danmaigoro
- Department of Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor 434000, Selangor, Malaysia
| | - Alhaji Zubair Jaji
- Department of Preclinical Science, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Selangor 434000, Selangor, Malaysia
| | - Zuki Abu Bakar
- Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
36
|
Effect of non-adhering dressings on promotion of fibroblast proliferation and wound healing in vitro. Sci Rep 2019; 9:4320. [PMID: 30867534 PMCID: PMC6416289 DOI: 10.1038/s41598-019-40921-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 02/21/2019] [Indexed: 12/15/2022] Open
Abstract
Non-adhering dressings are commonly used during granulation, tissue formation, and re-epithelialization. Elucidating cytotoxic effects and influence on proliferation/migration capacity of cells like fibroblasts is of interest. Dressings’ effects were investigated by comprehensive in vitro approach: (1) MTT assay measuring cell viability after direct contact, (2) ATP assay determining effects on cell proliferation, and (3) scratch wound assay featuring an in vitro wound healing model. One cotton-based dressing with vaseline (vas) was included in the study and four polyester dressings containing vas and technology-lipido-colloid matrix (TLC), carboxymethylcellulose (CMC), hydrocolloid (HC), or glycerin (gly) as additives. A polyamide dressing with vas + CMC and three silicone-based dressings (AT, CC, M) were tested. Polyester + vas + CMC did not negatively affect cell viability or proliferation but it was found that fibroblast layers appeared more irregular with decreased F-actin network structure and tubulin density possibly leading to hampered scratch closure. Silicone AT, polyester + gly and polyamide + vas + CMC caused distinct cell damage. The latter two further reduced cell viability, proliferation and scratch healing. From the overall results, it can be concluded that cotton + vas, polyester + TLC, polyester + vas + HC and the silicone dressings CC and M have the potential to prevent damage of newly formed tissue during dressing changes and positively influence wound healing.
Collapse
|
37
|
Emami F, Banstola A, Vatanara A, Lee S, Kim JO, Jeong JH, Yook S. Doxorubicin and Anti-PD-L1 Antibody Conjugated Gold Nanoparticles for Colorectal Cancer Photochemotherapy. Mol Pharm 2019; 16:1184-1199. [PMID: 30698975 DOI: 10.1021/acs.molpharmaceut.8b01157] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death worldwide. The prognosis and overall survival of CRC are known to be significantly correlated with the overexpression of PD-L1. Since combination therapies can significantly improve therapeutic efficacy, we constructed doxorubicin (DOX) conjugated and anti-PD-L1 targeting gold nanoparticles (PD-L1-AuNP-DOX) for the targeted chemo-photothermal therapy of CRC. DOX and anti-PD-L1 antibody were conjugated to the α-terminal end group of lipoic acid polyethylene glycol N-hydroxysuccinimide (LA-PEG-NHS) using an amide linkage, and PD-L1-AuNP-DOX was constructed by linking LA-PEG-DOX, LA-PEG-PD-L1, and a short PEG chain on the surface of AuNP using thiol-Au covalent bonds. Physicochemical characterizations and biological studies of PD-L1-AuNP-DOX were performed in the presence of near-infrared (NIR) irradiation (biologic studies were conducted using cellular uptake, apoptosis, and cell cycle assays in CT-26 cells). PD-L1-AuNP-DOX (40.0 ± 3.1 nm) was successfully constructed and facilitated the efficient intracellular uptake of DOX as evidenced by pronounced apoptotic effects (66.0%) in CT-26 cells. PD-L1-AuNP-DOX treatment plus NIR irradiation significantly and synergistically suppressed the in vitro proliferation of CT-26 cells by increasing apoptosis and cell cycle arrest. The study demonstrates that PD-L1-AuNP-DOX in combination with synergistic targeted chemo-photothermal therapy has a considerable potential for the treatment of localized CRC.
Collapse
Affiliation(s)
- Fakhrossadat Emami
- College of Pharmacy , Tehran University of Medical Science , Tehran , Iran
| | - Asmita Banstola
- College of Pharmacy , Keimyung University , Daegu 42601 , Republic of Korea
| | - Alireza Vatanara
- College of Pharmacy , Tehran University of Medical Science , Tehran , Iran
| | - Sooyeon Lee
- College of Pharmacy , Keimyung University , Daegu 42601 , Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy , Yeungnam University , Gyeongsan , Gyeongbuk 38541 , Republic of Korea
| | - Jee-Heon Jeong
- College of Pharmacy , Yeungnam University , Gyeongsan , Gyeongbuk 38541 , Republic of Korea
| | - Simmyung Yook
- College of Pharmacy , Keimyung University , Daegu 42601 , Republic of Korea
| |
Collapse
|
38
|
Guazuma ulmifolia Lam. Decreases Oxidative Stress in Blood Cells and Prevents Doxorubicin-Induced Cardiotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2935051. [PMID: 30050650 PMCID: PMC6046128 DOI: 10.1155/2018/2935051] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/19/2018] [Accepted: 04/01/2018] [Indexed: 02/07/2023]
Abstract
Doxorubicin (DOX) is an efficient chemotherapeutic agent, but its clinical application is limited by its cardiotoxicity associated with increased oxidative stress. Thus, the combination of DOX and antioxidants has been encouraged. In this study, we evaluated (I) the chemical composition and antioxidant capacity of aqueous extracts from Guazuma ulmifolia stem bark (GUEsb) and leaves (GUEl) in 2,2-diphenyl-1-picrylhydrazyl (DPPH) free radical scavenging, 2,2′-azobis(2-amidinopropane) dihydrochloride- (AAPH-) or DOX-induced lipid peroxidation inhibition in human blood cells, and intracellular reactive oxygen species (ROS) quantification using the fluorescent probe dichloro-dihydro-fluorescein diacetate (DCFH-DA) in K562 erythroleukemia cells incubated with GUEsb and stimulated with hydrogen peroxide; (II) the viability of K562 cells and human leukocytes treated with GUEsb in the absence or presence of DOX using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay; (III) the acute toxicity of GUEsb; and (IV) the cardioprotective effect of GUEsb in C57Bl/6 mice treated with DOX. The chemical composition indicated the presence of flavan-3-ol derivatives and condensed tannins in GUEsb and glycosylated flavonoids in GUEl. GUEsb and GUEl showed free-radical scavenging antioxidant activity, antihemolytic activity, and AAPH- as well as DOX-induced malondialdehyde content reduction in human erythrocytes. Based on its higher antioxidant potential, GUEsb was selected and subsequently showed intracellular ROS reduction without impairing the chemotherapeutic activity of DOX in K562 cells or inducing leukocyte cell death, but protected them against DOX-induced cell death. Yet, GUEsb did not show in vivo acute toxicity, and it prevented MDA generation in the cardiac tissue of DOX-treated mice, thus demonstrating its cardioprotective effect. Taken together, the results show that GUEsb and GUEl are natural alternatives to treat diseases associated with oxidative stress and that, in particular, GUEsb may play an adjuvant role in DOX chemotherapy.
Collapse
|
39
|
Bram Ednersson S, Stenson M, Stern M, Enblad G, Fagman H, Nilsson-Ehle H, Hasselblom S, Andersson PO. Expression of ribosomal and actin network proteins and immunochemotherapy resistance in diffuse large B cell lymphoma patients. Br J Haematol 2018; 181:770-781. [PMID: 29767447 DOI: 10.1111/bjh.15259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/05/2018] [Indexed: 10/25/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL) patients with early relapse or refractory disease have a very poor outcome. Immunochemotherapy resistance will probably, also in the era of targeted drugs, remain the major cause of treatment failure. We used proteomic mass spectrometry to analyse the global protein expression of micro-dissected formalin-fixed paraffin-embedded tumour tissues from 97 DLBCL patients: 44 with primary refractory disease or relapse within 1 year from diagnosis (REF/REL), and 53 who were progression-free more than 5 years after diagnosis (CURED). We identified 2127 proteins: 442 were found in all patients and 102 were differentially expressed. Sixty-five proteins were overexpressed in REF/REL patients, of which 46 were ribosomal proteins (RPs) compared with 2 of the 37 overexpressed proteins in CURED patients (P = 7·6 × 10-10 ). Twenty of 37 overexpressed proteins in CURED patients were associated with actin regulation, compared with 1 of 65 in REF/REL patients (P = 1·4 × 10-9 ). Immunohistochemical staining showed higher expression of RPS5 and RPL17 in REF/REL patients while MARCKS-like protein, belonging to the actin network, was more highly expressed in CURED patients. Even though functional studies aimed at individual proteins and protein interactions to evaluate potential clinical effect are needed, our findings suggest new mechanisms behind immunochemotherapy resistance in DLBCL.
Collapse
Affiliation(s)
- Susanne Bram Ednersson
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Martin Stenson
- Section of Haematology, Department of Medicine, Kungälvs Hospital, Kungälv, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mimmie Stern
- Section of Haematology, Department of Medicine, South Älvsborg Hospital, Borås, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics and Pathology/Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden
| | - Henrik Fagman
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Herman Nilsson-Ehle
- Section of Haematology and Coagulation, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sverker Hasselblom
- Department of Research, Development and Education, Region Halland, Halmstad, Sweden
| | - Per-Ola Andersson
- Section of Haematology, Department of Medicine, South Älvsborg Hospital, Borås, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
40
|
Shi J, Surma M, Wei L. Disruption of ROCK1 gene restores autophagic flux and mitigates doxorubicin-induced cardiotoxicity. Oncotarget 2018; 9:12995-13008. [PMID: 29560126 PMCID: PMC5849190 DOI: 10.18632/oncotarget.24457] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 02/03/2018] [Indexed: 12/25/2022] Open
Abstract
Doxorubicin is among the essential medicines with a wide antitumor spectrum, but its clinical application is limited by its cardiotoxicity. We recently discovered that ROCK1 is a key molecule in mediating cardiac remodeling in response to various stresses. To determine the roles of ROCK1 in doxorubicin cardiotoxicity, we gave three doses of doxorubicin injections to wild type (WT) and ROCK1−/− mice with one week intervals between treatments, the cumulative dose being 24 mg/kg. ROCK1−/− mice exhibited preserved cardiac function, reduced apoptosis, autophagy and fibrosis compared to the WT mice. To further determine the cellular mechanisms, we have examined the role of ROCK1 in cardiomyocytes using cardiomyocyte-specific knockout mice, MHC-Cre/ROCK1fl/fl, which partially reproduced the cardioprotective characteristics of ROCK1−/− mice, indicating that ROCK1 in both cardiomyocytes and non-cardiomyocytes mediates doxorubicin cardiotoxicity. To elucidate the molecular mechanisms, a detailed time course study after a single doxorubicin injection at 10 mg/kg was performed in ROCK1−/− and MHC-Cre/ROCK1fl/fl mice. The molecular analysis revealed that both ROCK1−/− and MHC-Cre/ROCK1fl/fl hearts exhibited significant reduction of doxorubicin-induced early responses including increased apoptotic (Bax) and autophagic (p62/SQSTM1 and LC3-II) markers, associated with reduced Beclin 1 phosphorylation on Thr119, supporting reduced Beclin 1-mediated autophagy initiation due to increased association of Beclin 1 with Bcl 2 or Bcl-XL in these hearts compared to the WT or ROCK1fl/fl mice. These results support that ROCK1 deficiency is cardioprotective against doxorubicin-induced cardiotoxicity at least in part through reducing Beclin 1-mediated autophagy initiation in cardiomyocytes and restoring autophagic flux to ameliorate doxorubicin cardiotoxicity.
Collapse
Affiliation(s)
- Jianjian Shi
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, USA
| | - Michelle Surma
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, USA
| | - Lei Wei
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, Indianapolis, Indiana, USA.,Department of Cellular and Integrative Physiology, Indiana University, School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
41
|
The Protective Effect of Total Flavones from Rhododendron simsii Planch. on Myocardial Ischemia/Reperfusion Injury and Its Underlying Mechanism. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018. [PMID: 29541143 PMCID: PMC5818901 DOI: 10.1155/2018/6139372] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Objectives Total flavones from Rhododendron simsii Planch. (TFR) are the effective part extracted from the flowers of Rhododendron simsii Planch. and have obvious protective effects against cerebral ischemic or myocardial injuries in rabbits and rats. However, their mechanism of cardioprotection is still unrevealed. Therefore, the present study was designed to investigate the effect of TFR on myocardial I/R injury and the underlying mechanism. Methods TFR groups were treated by gavage once a day for 3 days at a dose of 20, 40, and 80 mg/kg, respectively, and then the model of myocardial I/R injury was established. Myocardial infarction, ST-segment elevation, and the expression of UTR, ROCK1, ROCK2, and p-MLC protein in rat myocardium were determined at 90 min after reperfusion. UTR siRNA in vivo transfection and competition binding assay method were used to study the relationship between the protective effect of TFR and UTR. Results The expression of UTR protein markedly decreased in myocardium of UTR siRNA transfection group rats. TFR could significantly reduce the infarct size and inhibit the increase of RhoA activity and ROCK1, ROCK2, and p-MLC protein expressions both in WT and UTR knockdown rats. The reducing rate of TFR in myocardial infarction area, RhoA activity, and ROCK1, ROCK2, and p-MLC protein expressions in UTR knockdown rats decreased markedly compared with that in WT rats. In addition, TFR had no obvious effect on the increase of ΣST in UTR knockdown rats in comparison with that in model group. In particular, TFR could significantly inhibit the combination of [125I]-hu-II and UTR, and IC50 was 0.854 mg/l. Conclusions The results indicate that the protective effect of TFR on I/R injury may be correlated with its blocking UTR and the subsequent inhibition of RhoA/ROCK signaling pathway.
Collapse
|
42
|
Korean red ginseng attenuates doxorubicin-induced testicular dysfunction in rats by modulating inflammatory, oxidative, and autophagy responses. J Funct Foods 2018. [DOI: 10.1016/j.jff.2017.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
|
43
|
Polonchuk L, Chabria M, Badi L, Hoflack JC, Figtree G, Davies MJ, Gentile C. Cardiac spheroids as promising in vitro models to study the human heart microenvironment. Sci Rep 2017; 7:7005. [PMID: 28765558 PMCID: PMC5539326 DOI: 10.1038/s41598-017-06385-8] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 06/26/2017] [Indexed: 12/12/2022] Open
Abstract
Three-dimensional in vitro cell systems are a promising alternative to animals to study cardiac biology and disease. We have generated three-dimensional in vitro models of the human heart ("cardiac spheroids", CSs) by co-culturing human primary or iPSC-derived cardiomyocytes, endothelial cells and fibroblasts at ratios approximating those present in vivo. The cellular organisation, extracellular matrix and microvascular network mimic human heart tissue. These spheroids have been employed to investigate the dose-limiting cardiotoxicity of the common anti-cancer drug doxorubicin. Viability/cytotoxicity assays indicate dose-dependent cytotoxic effects, which are inhibited by the nitric oxide synthase (NOS) inhibitor L-NIO, and genetic inhibition of endothelial NOS, implicating peroxynitrous acid as a key damaging agent. These data indicate that CSs mimic important features of human heart morphology, biochemistry and pharmacology in vitro, offering a promising alternative to animals and standard cell cultures with regard to mechanistic insights and prediction of toxic effects in human heart tissue.
Collapse
Affiliation(s)
- Liudmila Polonchuk
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Mamta Chabria
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Laura Badi
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Jean-Christophe Hoflack
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, 4070, Switzerland
| | - Gemma Figtree
- Sydney Medical School, University of Sydney, Sydney, 2000, Australia
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Carmine Gentile
- Sydney Medical School, University of Sydney, Sydney, 2000, Australia.
- Heart Research Institute, Newtown, 2041, Australia.
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA.
| |
Collapse
|
44
|
Bai G, Li Y, Chu HK, Wang K, Tan Q, Xiong J, Sun D. Characterization of biomechanical properties of cells through dielectrophoresis-based cell stretching and actin cytoskeleton modeling. Biomed Eng Online 2017; 16:41. [PMID: 28376803 PMCID: PMC5381122 DOI: 10.1186/s12938-017-0329-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 03/16/2017] [Indexed: 02/02/2023] Open
Abstract
Background Cytoskeleton is a highly dynamic network that helps to maintain the rigidity of a cell, and the mechanical properties of a cell are closely related to many cellular functions. This paper presents a new method to probe and
characterize cell mechanical properties through dielectrophoresis (DEP)-based cell stretching manipulation and actin cytoskeleton modeling. Methods Leukemia NB4 cells were used as cell line, and changes in their biological properties were examined after chemotherapy treatment with doxorubicin (DOX). DEP-integrated microfluidic chip was utilized as a low-cost and efficient tool to study the deformability of cells. DEP forces used in cell stretching were first evaluated through computer simulation, and the results were compared with modeling equations and with the results of optical stretching (OT) experiments. Structural parameters were then extracted by fitting the experimental data into the actin cytoskeleton model, and the underlying mechanical properties of the cells were subsequently characterized. Results The DEP forces generated under different voltage inputs were calculated and the results from different approaches demonstrate good approximations to the force estimation. Both DEP and OT stretching experiments confirmed that DOX-treated NB4 cells were stiffer than the untreated cells. The structural parameters extracted from the model and the confocal images indicated significant change in actin network after DOX treatment. Conclusion The proposed DEP method combined with actin cytoskeleton modeling is a simple engineering tool to characterize the mechanical properties of cells.
Collapse
Affiliation(s)
- Guohua Bai
- Key Laboratory of Instrumentation Science and Dynamic Measurement, Ministry of Education, North University of China, Room 418, Building No. 14, No. 3 Xueyuan Road, Taiyuan, 030051, Shanxi, China
| | - Ying Li
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR of China
| | - Henry K Chu
- Department of Mechanical Engineering, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, SAR of China
| | - Kaiqun Wang
- Department of Biomedical Engineering, College of Mechanics, Taiyuan University of Technology, No. 79, West Yingze Street, Taiyuan, 030024, Shanxi, China
| | - Qiulin Tan
- Key Laboratory of Instrumentation Science and Dynamic Measurement, Ministry of Education, North University of China, Room 418, Building No. 14, No. 3 Xueyuan Road, Taiyuan, 030051, Shanxi, China
| | - Jijun Xiong
- Key Laboratory of Instrumentation Science and Dynamic Measurement, Ministry of Education, North University of China, Room 418, Building No. 14, No. 3 Xueyuan Road, Taiyuan, 030051, Shanxi, China
| | - Dong Sun
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong, SAR of China.
| |
Collapse
|
45
|
Pi J, Huang L, Yang F, Jiang J, Jin H, Liu J, Su X, Wu A, Cai H, Yang P, Cai J. Atomic force microscopy study of ionomycin-induced degranulation in RBL-2H3 cells. SCANNING 2016; 38:525-534. [PMID: 26840764 DOI: 10.1002/sca.21291] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/22/2015] [Indexed: 06/05/2023]
Abstract
Mast cell degranulation is the typical anaphylaxis process of mast cells associated with the release of cytokines, eicosanoids and their secretory granules, which play very important roles in the allergic inflammatory response of the human body upon anaphylactogen stimulation. The calcium ionophore ionomycin is widely used as a degranulation induction agent for mast cell degranulation studies. In the present work, ionomycin-induced degranulation of RBL-2H3 basophilic leukemia cell line cells was investigated in vitro by high resolution atomic force microscopy (AFM). Ionomycin, which could increase the intracellular free Ca2+ level and β-Hexosaminidase release, was found to induce the formation of a kind of peculiar vesicles in the cytoplasm area of RBL-2H3 cells. Those vesicles induced by ionomycin would desintegrate to release a larger amount of granules surrounding RBL-2H3 cells by the controlling of F-actin. These results provide the precise morphological information of ionomycin-induced mast cell degranulation at nanoscale, which could benefit our understanding of ionomycin-induced mast cell anaphylaxis model and also validate the applicability of AFM for the detection of allergic inflammatory response in mast cells. SCANNING 38:525-534, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jiang Pi
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Lufen Huang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Fen Yang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jinhuan Jiang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Hua Jin
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jianxin Liu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
- Department of Pharmacology, Hunan University of Medicine, Huaihua, China
| | - Xiaohui Su
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Anguo Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Huaihong Cai
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Peihui Yang
- Department of Pharmacology, Hunan University of Medicine, Huaihua, China
| | - Jiye Cai
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
- Department of Chemistry, Jinan University, Guangzhou, China
| |
Collapse
|
46
|
Zhang T, Shao Y, Chu TY, Huang HS, Liou YL, Li Q, Zhou H. MiR-135a and MRP1 play pivotal roles in the selective lethality of phenethyl isothiocyanate to malignant glioma cells. Am J Cancer Res 2016; 6:957-972. [PMID: 27293991 PMCID: PMC4889712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 03/02/2016] [Indexed: 06/06/2023] Open
Abstract
Amounting evidence has demonstrated that phenethyl isothiocyanate (PEITC) is a strong inducer of reactive oxygen species (ROS) and functions as a selective killer to various human cancer cells. However, it remains obscure whether PEITC has potential selective lethality to malignant glioma cells. Thus in this study, we performed multiple analysis such as MTT assay, Hoechst 33258 staining, flow cytometry, foci formation, RT-PCR, Western blot, and transfection to explore the selective lethality of PEITC to malignant glioma cells and the underlying mechanisms. We found that PEITC induced a selective apoptosis and suppressed tumorigenicity and migration of malignant glioma cells. Furthermore, we found PEITC significantly induced GSH depletion, ROS production, caspase-9 and caspase-3 activation, and miR-135a upregulation in malignant glioma cells but not in normal cells. Moreover, PEITC activated the miR-135a-mitochondria dependent apoptosis pathway as demonstrated by downregulation of STAT6, SMAD5 and Bcl-xl while upregulation of Bax expression and Cytochrome-C release in malignant glioma cell lines but not in the immortalized human normal glial HEB cells. Correspondingly, the above PEITC-induced activation of the ROS-MiR-135a-Mitochondria dependent apoptosis pathways in malignant glioma was attenuated by pre-transfection with miR-135a inhibitor, pre-treatment with multidrug resistance-associated protein 1 (MRP1) inhibitor Sch B, or combination with glutathione (GSH). These results revealed that PEITC selectively induced apoptosis of malignant glioma cells through MRP1-mediated export of GSH to activate ROS-MiR-135a-Mitochondria dependent apoptosis pathway, suggesting a potential application of PEITC for treating glioma.
Collapse
Affiliation(s)
- Taolan Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South UniversityChangsha 410008, P. R. China
- Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of PharmacogeneticsChangsha 410078, P. R. China
| | - Yingying Shao
- Institute of Life Sciences, Chongqing Medical University1 Yixueyuan Rd, Yuzhong District, Chongqing 400016, P. R. China
| | - Tang-Yuan Chu
- Institute of Medical Sciences, Tzu Chi UniversityHualien 970, Taiwan
| | - Hsuan-Shun Huang
- Institute of Medical Sciences, Tzu Chi UniversityHualien 970, Taiwan
| | - Yu-Ligh Liou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South UniversityChangsha 410008, P. R. China
- Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of PharmacogeneticsChangsha 410078, P. R. China
- IStat Biomedical Co. Ltd.Taipei, Taiwan
| | - Qing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South UniversityChangsha 410008, P. R. China
- Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of PharmacogeneticsChangsha 410078, P. R. China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South UniversityChangsha 410008, P. R. China
- Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of PharmacogeneticsChangsha 410078, P. R. China
| |
Collapse
|
47
|
Wei L, Surma M, Shi S, Lambert-Cheatham N, Shi J. Novel Insights into the Roles of Rho Kinase in Cancer. Arch Immunol Ther Exp (Warsz) 2016; 64:259-78. [PMID: 26725045 PMCID: PMC4930737 DOI: 10.1007/s00005-015-0382-6] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 11/24/2015] [Indexed: 12/12/2022]
Abstract
Rho-associated coiled-coil kinase (ROCK) is a major downstream effector of the small GTPase RhoA. The ROCK family, consisting of ROCK1 and ROCK2, plays a central role in the organization of the actin cytoskeleton, and is involved in a wide range of fundamental cellular functions such as contraction, adhesion, migration, proliferation, and apoptosis. Since the discovery of effective inhibitors such as fasudil and Y27632, the biological roles of ROCK have been extensively explored in numerous diseases, including cancer. Accumulating evidence supports the concept that ROCK plays important roles in tumor development and progression through regulating many key cellular functions associated with malignancy, including tumorigenicity, tumor growth, metastasis, angiogenesis, tumor cell apoptosis/survival and chemoresistance as well. This review focuses on the new advances of the most recent 5 years from the studies on the roles of ROCK in cancer development and progression; the discussion is mainly focused on the potential value of ROCK inhibitors in cancer therapy.
Collapse
Affiliation(s)
- Lei Wei
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA. .,Department of Cellular and Integrative Physiology, Indiana University, School of Medicine, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| | - Michelle Surma
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA
| | - Stephanie Shi
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA
| | - Nathan Lambert-Cheatham
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA
| | - Jianjian Shi
- Riley Heart Research Center, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University, School of Medicine, R4 Building, Room 332, 1044 West Walnut Street, Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|