1
|
Ashrafi P, Sari S, Javani Jouni F, Zafari J, Asgari F. Potentiated Effects of Photobiomodulation and Celecoxib on the Epithelial-Mesenchymal Transition Signaling of E-Cadherin, N-Cadherin, α-SMA in Breast Cancer Cells, MCF7, and MDA-MB-231. Photobiomodul Photomed Laser Surg 2025; 43:115-123. [PMID: 39992209 DOI: 10.1089/photob.2024.0155] [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: 02/25/2025] Open
Abstract
Background and Objective: Breast cancer (BC) is one of the most common cancers among women, with a high potential for metastasis. The epithelial-mesenchymal transition (EMT) is crucial in the invasion and metastasis of cancer cells. This research was designed to examine the efficacy of photobiomodulation therapy in combination with celecoxib in inhibiting the EMT process. We also analyzed the changes in the expression of E-cadherin, N-cadherin, and α-SMA genes in BC cell lines MCF-7 and MDA-MB-231. Material and Methods: In this study, the IC50 of celecoxib was first determined using the 3-(4,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide assay for both cell lines. The cells were then treated with celecoxib, laser irradiation, and their combination. A migration assay was performed to evaluate the cell migration. Real-time polymerase chain reaction also assessed the changes in the expression of the abovementioned genes. Results: A combination of celecoxib and laser therapy significantly reduced the migration of cancer cells. Additionally, the potentiated effect of the combined therapy altered the expression levels of the aforementioned genes, indicating the potential role of the combination treatment in regulating EMT. Conclusions: Our research discloses that combining laser therapy with celecoxib could serve as an effective therapeutic approach to inhibit BC invasion and metastasis by targeting the EMT process and decelerating disease progression. Further investigations are essential to validate these results in clinical environments.
Collapse
Affiliation(s)
- Parisa Ashrafi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Soyar Sari
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Javani Jouni
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Jaber Zafari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Asgari
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Ebrahimi S, Khaleghi Ghadiri M, Stummer W, Gorji A. Enhancing 5-ALA-PDT efficacy against resistant tumor cells: Strategies and advances. Life Sci 2024; 351:122808. [PMID: 38852796 DOI: 10.1016/j.lfs.2024.122808] [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: 04/04/2024] [Revised: 05/20/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
As a precursor of protoporphyrin IX (PpIX), an endogenous pro-apoptotic and fluorescent molecule, 5-Aminolevulinic acid (5-ALA) has gained substantial attention for its potential in fluorescence-guided surgery as well as photodynamic therapy (PDT). Moreover, 5-ALA-PDT has been suggested as a promising chemo-radio sensitization therapy for various cancers. However, insufficient 5-ALA-induced PpIX fluorescence and the induction of multiple resistance mechanisms may hinder the 5-ALA-PDT clinical outcome. Reduced efficacy and resistance to 5-ALA-PDT can result from genomic alterations, tumor heterogeneity, hypoxia, activation of pathways related to cell surveillance, production of nitric oxide, and most importantly, deregulated 5-ALA transporter proteins and heme biosynthesis enzymes. Understanding the resistance regulatory mechanisms of 5-ALA-PDT may allow the development of effective personalized cancer therapy. Here, we described the mechanisms underlying resistance to 5-ALA-PTD across various tumor types and explored potential strategies to overcome this resistance. Furthermore, we discussed future approaches that may enhance the efficacy of treatments using 5-ALA-PDT.
Collapse
Affiliation(s)
- Safieh Ebrahimi
- Epilepsy Research Center, Münster University, 48149 Münster, Germany; Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran
| | | | - Walter Stummer
- Department of Neurosurgery, Münster University, 48149 Münster, Germany
| | - Ali Gorji
- Epilepsy Research Center, Münster University, 48149 Münster, Germany; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran 1996835911, Iran; Neuroscience Research Center, Mashhad University of Medical Sciences, 9177948564 Mashhad, Iran.
| |
Collapse
|
3
|
Udrea AM, Dinache A, Staicu A, Avram S. Target Prediction of 5,10,15,20-Tetrakis(4'-Sulfonatophenyl)-Porphyrin Using Molecular Docking. Pharmaceutics 2022; 14:2390. [PMID: 36365208 PMCID: PMC9692331 DOI: 10.3390/pharmaceutics14112390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 09/10/2024] Open
Abstract
Photodynamic therapy has the potential to be a new and effective cancer treatment. Even if in vitro and in vivo research show promise, the molecular mechanism remains unclear. In this study, molecular docking simulations predict the binding affinity of the 5,10,15,20-tetrakis(4'-sulfonatophenyl)-porphyrin tetraammonium photosensitizer on several potential targets in photodynamic treatment. Our results indicate that this photosensitizer binds to several receptor targets, including B-cell lymphoma 2 (BCL-2) and other related proteins BCL-xL, MCL-1, or A1. The binding affinity of the porphyrin derivative with human serum albumin was determined using UV-vis absorption spectroscopy and predicted using molecular docking. We conclude that the studied porphyrin photosensitizer binds to human serum albumin and may inhibit the cancer cell line through its interactions with HIS and MET AA residues from BCL-2, MCL-1, and β-catenin receptors or through its low estimated free energy of binding when interacting with A1 and BCL-B receptors.
Collapse
Affiliation(s)
- Ana-Maria Udrea
- Laser Department, National Institute for Laser, Plasma and Radiation Physics, Atomistilor 409, 077125 Magurele, Romania
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| | - Andra Dinache
- Laser Department, National Institute for Laser, Plasma and Radiation Physics, Atomistilor 409, 077125 Magurele, Romania
| | - Angela Staicu
- Laser Department, National Institute for Laser, Plasma and Radiation Physics, Atomistilor 409, 077125 Magurele, Romania
| | - Speranta Avram
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| |
Collapse
|
4
|
Quiñones OG, Pierre MBR. Cutaneous Application of Celecoxib for Inflammatory and Cancer Diseases. Curr Cancer Drug Targets 2020; 19:5-16. [PMID: 29714143 DOI: 10.2174/1568009618666180430125201] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/31/2018] [Accepted: 03/03/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drugs (NSAIDs) and particularly selective cyclooxygenase-2 (COX-2) inhibitors such as celecoxib (Cxb) are considered promising cancer chemopreventive for colon, breast, prostate, lung, and skin cancers. However, the clinical application to the prevention is limited by concerns about safety, potential to serious toxicity (mainly for healthy individuals), efficacy and optimal treatment regimen. Cxb exhibits advantages as potent antiinflammatory and gastrointestinal tolerance compared with conventional NSAID's. Recent researches suggest that dermatological formulations of Cxb are more suitable than oral administration in the treatment of cutaneous disease, including skin cancer. To date, optimism has been growing regarding the exploration of the topical application of Cxb (in the prevention of skin cancers and treatment of cutaneous inflammation) or transdermal route reducing risks of systemic side effects. OBJECTIVE This paper briefly summarizes our current knowledge of the development of the cutaneous formulations or delivery systems for Cxb as anti-inflammatory drug (for topical or transdermal application) as well its chemopreventive properties focused on skin cancer. CONCLUSION New perspectives emerge from the growing knowledge, bringing innovative techniques combining the action of Cxb with other substances or agents which act in a different way, but complementary, increasing the efficacy and minimizing toxicity.
Collapse
Affiliation(s)
- Oliesia Gonzalez Quiñones
- School of Pharmacy, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, 21.941.902, Rio de Janeiro, RJ, Brazil
| | - Maria Bernadete Riemma Pierre
- School of Pharmacy, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho 373, 21.941.902, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
5
|
Kraus D, Palasuberniam P, Chen B. Therapeutic Enhancement of Verteporfin-mediated Photodynamic Therapy by mTOR Inhibitors. Photochem Photobiol 2019; 96:358-364. [PMID: 31769520 DOI: 10.1111/php.13187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/31/2019] [Indexed: 01/17/2023]
Abstract
Photodynamic therapy (PDT) with photosensitizer verteporfin is a clinically approved vascular disrupting modality that is currently in clinical trial for cancer treatment. In this study, we evaluated PDT in combination with either mTORC1 inhibitor rapamycin or mTORC1/C2 dual inhibitor AZD2014 for therapeutic enhancement in SVEC endothelial cells. Verteporfin-PDT alone induced cell apoptosis by activating the intrinsic apoptotic pathway. However, it increased the expression of anti-apoptotic protein MCL-1 and the phosphorylation of S6, a downstream molecule of mTOR signaling. In contrast, mTOR inhibitors rapamycin and AZD2014 did not induce apoptosis in SVEC cells. They suppressed MCL-1 expression and S6 phosphorylation and imposed a potent inhibition on cell proliferation. PDT in combination with mTOR inhibitors activated the intrinsic apoptotic pathway and resulted in increased apoptosis. Combination treatments also led to sustained inhibition of cell proliferation. Although AZD2014 was more effective for cell growth inhibition and PDT enhancement than rapamycin at the higher concentrations examined in the study, both inhibitors effectively enhanced PDT response, suggesting that inhibition of mTORC1 is crucial for PDT enhancement. Our results indicate that mTOR inhibitors mechanistically cooperate with PDT for enhanced cell death and sustained growth inhibition, supporting a combination approach for therapeutic enhancement.
Collapse
Affiliation(s)
- Daniel Kraus
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA
| | - Pratheeba Palasuberniam
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA
| | - Bin Chen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA.,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
6
|
The molecular targets of diclofenac differs from ibuprofen to induce apoptosis and epithelial mesenchymal transition due to alternation on oxidative stress management p53 independently in PC3 prostate cancer cells. Prostate Int 2019; 7:156-165. [PMID: 31970141 PMCID: PMC6962753 DOI: 10.1016/j.prnil.2019.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 08/19/2019] [Accepted: 09/25/2019] [Indexed: 01/16/2023] Open
Abstract
Background Prostate cancer is the most common type of cancer among men. Studies showed that the regular use of nonsteroidal antiinflammatory drugs might reduce disease progression risk for prostate cancer patients with prostate cancer. We evaluated the effects of ectopic expression of p53 on the biological functions of ibuprofen and diclofenac. Materials and methods For this purpose, We investigated cell death decision pathways related to survival and aggressive cellular phenotypes such as extrinsic/intrinsic apoptosis decision, Protein Kinase B/ Forkhead box O (AKT/FoxO) axis, mitogen-activated protein kinases (MAPKs), reactive oxygen species (ROS) generation, and EMT (epithelial mesenchymal transition) in wild type and p53 + PC3 prostate cancer cells. Results and Conclusions Ibuprofen (1 mM) and diclofenac (250 μM) effectively induced cell cycle arrest and led to apoptosis via modulating both extrinsic and intrinsic pathways. However, diclofenac was the only drug to generate ROS intermediates. Diclofenac triggered a typical EMT process with downregulated E-cadherin and upregulated N-cadherin, vimentin, and Snail in PC3 cells, regardless of p53 expression. In conclusion, although both drugs are effective on cell death mechanism, only diclofenac caused EMT because of increased ROS generation independent of p53. On the other hand, ibuprofen could inhibit metastasis via upregulating E-cadherin. The biological targets of both nonsteroidal antiinflammatory drugs are different to highlight their role in cell survival and death axis.
Collapse
|
7
|
Halatsch M, Kast RE, Dwucet A, Hlavac M, Heiland T, Westhoff M, Debatin K, Wirtz CR, Siegelin MD, Karpel‐Massler G. Bcl-2/Bcl-xL inhibition predominantly synergistically enhances the anti-neoplastic activity of a low-dose CUSP9 repurposed drug regime against glioblastoma. Br J Pharmacol 2019; 176:3681-3694. [PMID: 31222722 PMCID: PMC6715605 DOI: 10.1111/bph.14773] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/27/2019] [Accepted: 06/04/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Drug repurposing represents a promising approach to safely accelerate the clinical application of therapeutics with anti-cancer activity. In this study, we examined whether inhibition of the anti-apoptotic Bcl-2 family proteins Bcl-2 and Bcl-xL enhances the biological effects of the repurposed CUSP9 regimen in an in vitro setting of glioblastoma. EXPERIMENTAL APPROACH We applied 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assays to assess cellular proliferation. Annexin V/propidium iodide and tetramethylrhodamine, ethyl ester staining were used to examine apoptosis. Western blotting, RT-PCR, and specific knockdown experiments using siRNA were employed to examine molecular mechanisms of action. KEY RESULTS Bcl-2/Bcl-xL inhibition exerted synergistic anti-proliferative effects across established, primary cultured, and stem-like glioblastoma cells when combined with CUSP9 which had been reduced to only one tenth of its proposed original concentration (CUSP9-LD). The combination treatment also led to enhanced apoptosis with loss of mitochondrial membrane potential and activation of caspases. On the molecular level, CUSP9-LD counteracted ABT263-mediated up-regulation of Mcl-1. Silencing of Mcl-1 enhanced ABT263-mediated apoptosis which indicates that down-regulation of Mcl-1 is crucial for the induction of cell death by the combination treatment. CONCLUSION AND IMPLICATIONS These data suggest that Bcl-2/Bcl-xL inhibition enhances the susceptibility of glioblastoma cells towards CUSP9, allowing dramatic dose reduction and potentially decreased toxicity when applied clinically. A clinical trial involving the original CUSP doses (CUSP9v3) is currently ongoing in our institution (NCT02770378). The Bcl-2/Bcl-xL inhibitor ABT263 is in clinical trials and might represent a valuable adjunct to the original CUSP.
Collapse
Affiliation(s)
| | | | - Annika Dwucet
- Department of NeurosurgeryUlm University Medical CenterUlmGermany
| | - Michal Hlavac
- Department of NeurosurgeryUlm University Medical CenterUlmGermany
| | - Tim Heiland
- Department of NeurosurgeryUlm University Medical CenterUlmGermany
| | - Mike‐Andrew Westhoff
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | - Klaus‐Michael Debatin
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | | | - Markus David Siegelin
- Department of Pathology and Cell BiologyColumbia University Medical CenterNew YorkNYUSA
| | | |
Collapse
|
8
|
Rosin FCP, Teixeira MG, Pelissari C, Corrêa L. Photodynamic Therapy Mediated by 5-aminolevulinic Acid Promotes the Upregulation and Modifies the Intracellular Expression of Surveillance Proteins in Oral Squamous Cell Carcinoma. Photochem Photobiol 2018; 95:635-643. [PMID: 30267573 DOI: 10.1111/php.13029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 09/09/2018] [Indexed: 12/22/2022]
Abstract
Expression of proteins related to cell surveillance has been described in tumors presenting resistance to photodynamic therapy (PDT). The aim of this study was to verify whether there was upregulation of proteins related to resistance in oral squamous cell carcinoma (OSCC) after PDT. OSCC was chemically induced in rats and treated after one cycle of PDT mediated by 5-aminolevulinic acid (5-ALA-PDT). Immunolabeling of p-NFκB, Bcl-2, survivin, iNOS, p-Akt, p-mTOR and cyclin D1 was performed after the treatment. There was increased expression of Bcl-2 (P = 0.008), iNOS (P = 0.020), p-Akt (P = 0.020) and p-mTOR (P = 0.010) by surviving neoplastic cells after PDT when compared to the control. In conclusion, after one cycle of 5-ALA-mediated PDT, Bcl-2, p-Akt, p-mTOR and iNOS were upregulated in neoplastic cells of OSCC, suggesting an activation of antiapoptosis and cell proliferation pathways. This fact must be considered in the establishment of PDT protocols for OSCC treatment, mainly those in which PDT will be combined with chemotherapy drugs targeted at the studied proteins.
Collapse
Affiliation(s)
| | | | - Cibele Pelissari
- Oral Pathology Department, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Luciana Corrêa
- General Pathology Department, School of Dentistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
9
|
Rosin FCP, Teixeira MG, Pelissari C, Corrêa L. Resistance of oral cancer cells to 5‐ALA‐mediated photodynamic therapy. J Cell Biochem 2018; 119:3554-3562. [DOI: 10.1002/jcb.26541] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/05/2017] [Indexed: 01/14/2023]
Affiliation(s)
- Flávia Cristina P. Rosin
- Pathology DepartmentSchool of DentistryUniversity of São Paulo. Av Prof Lineu PrestesSão PauloBrazil
| | - Marina Gabriela Teixeira
- Pathology DepartmentSchool of DentistryUniversity of São Paulo. Av Prof Lineu PrestesSão PauloBrazil
| | - Cibele Pelissari
- Pathology DepartmentSchool of DentistryUniversity of São Paulo. Av Prof Lineu PrestesSão PauloBrazil
| | - Luciana Corrêa
- Pathology DepartmentSchool of DentistryUniversity of São Paulo. Av Prof Lineu PrestesSão PauloBrazil
| |
Collapse
|
10
|
Kraus D, Palasuberniam P, Chen B. Targeting Phosphatidylinositol 3-Kinase Signaling Pathway for Therapeutic Enhancement of Vascular-Targeted Photodynamic Therapy. Mol Cancer Ther 2017; 16:2422-2431. [PMID: 28835385 DOI: 10.1158/1535-7163.mct-17-0326] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 07/10/2017] [Accepted: 08/07/2017] [Indexed: 11/16/2022]
Abstract
Vascular-targeted photodynamic therapy (PDT) selectively disrupts vascular function by inducing oxidative damages to the vasculature, particularly endothelial cells. Although effective tumor eradication and excellent safety profile are well demonstrated in both preclinical and clinical studies, incomplete vascular shutdown and angiogenesis are known to cause tumor recurrence after vascular-targeted PDT. We have explored therapeutic enhancement of vascular-targeted PDT with PI3K signaling pathway inhibitors because the activation of PI3K pathway was involved in promoting endothelial cell survival and proliferation after PDT. Here, three clinically relevant small-molecule inhibitors (BYL719, BKM120, and BEZ235) of the PI3K pathway were evaluated in combination with verteporfin-PDT. Although all three inhibitors were able to synergistically enhance PDT response in endothelial cells, PDT combined with dual PI3K/mTOR inhibitor BEZ235 exhibited the strongest synergism, followed in order by combinations with pan-PI3K inhibitor BKM120 and p110α isoform-selective inhibitor BYL719. Combination treatments of PDT and BEZ235 exhibited a cooperative inhibition of antiapoptotic Bcl-2 family protein Mcl-1 and induced more cell apoptosis than each treatment alone. In addition to increasing treatment lethality, BEZ235 combined with PDT effectively inhibited PI3K pathway activation and consequent endothelial cell proliferation after PDT alone, leading to a sustained growth inhibition. In the PC-3 prostate tumor model, combination treatments improved treatment outcomes by turning a temporary tumor regrowth delay induced by PDT alone to a more long-lasting treatment response. Our study strongly supports the combination of vascular-targeted PDT and PI3K pathway inhibitors, particularly mTOR inhibitors, for therapeutic enhancement. Mol Cancer Ther; 16(11); 2422-31. ©2017 AACR.
Collapse
Affiliation(s)
- Daniel Kraus
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, Pennsylvania
| | - Pratheeba Palasuberniam
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, Pennsylvania
| | - Bin Chen
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, Pennsylvania. .,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Kou J, Dou D, Yang L. Porphyrin photosensitizers in photodynamic therapy and its applications. Oncotarget 2017; 8:81591-81603. [PMID: 29113417 PMCID: PMC5655312 DOI: 10.18632/oncotarget.20189] [Citation(s) in RCA: 333] [Impact Index Per Article: 41.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/29/2017] [Indexed: 01/09/2023] Open
Abstract
In 1841, the extraction of hematoporphyrin from dried blood by removing iron marked the birth of the photosensitizer. The last twenty years has witnessed extensive research in the application of photodynamic therapy (PDT) in tumor-bearing (or other diseases) animal models and patients. The period has seen development of photosensitizers from the first to the third generation, and their evolution from simple to more complex entities. This review focuses on porphyrin photosensitizers and their effect on tumors, mediated via several pathways involved in cell necrosis, apoptosis or autophagic cell death, and the preventive and therapeutic application of PDT against atherosclerosis.
Collapse
Affiliation(s)
- Jiayuan Kou
- Department of Pathophysiology, Harbin Medical University, Harbin, PR China.,Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Dou Dou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, PR China
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University, Harbin, PR China
| |
Collapse
|
12
|
Jin G, Feng G, Qin W, Tang BZ, Liu B, Li K. Multifunctional organic nanoparticles with aggregation-induced emission (AIE) characteristics for targeted photodynamic therapy and RNA interference therapy. Chem Commun (Camb) 2016; 52:2752-5. [DOI: 10.1039/c5cc07818k] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
We report the design of AIE fluorogen (AIEgen)-based multifunctional organic nanoparticles with surface siRNA decoration for targeted photodynamic therapy and RNA interference therapy.
Collapse
Affiliation(s)
- Guorui Jin
- Institute of Materials Research and Engineering
- A*STAR
- Singapore
| | - Guangxue Feng
- Department of Chemical and Biomolecular Engineering
- National University of Singapore
- Singapore
| | - Wei Qin
- Department of Chemistry
- Institute for Advanced Study
- Institute of Molecular Functional Materials
- Division of Biomedical Engineering, Division of Life Science
- and State Key Laboratory of Molecular Neuroscience
| | - Ben Zhong Tang
- Department of Chemistry
- Institute for Advanced Study
- Institute of Molecular Functional Materials
- Division of Biomedical Engineering, Division of Life Science
- and State Key Laboratory of Molecular Neuroscience
| | - Bin Liu
- Institute of Materials Research and Engineering
- A*STAR
- Singapore
- Department of Chemical and Biomolecular Engineering
- National University of Singapore
| | - Kai Li
- Institute of Materials Research and Engineering
- A*STAR
- Singapore
| |
Collapse
|
13
|
Ghosh R, Alajbegovic A, Gomes AV. NSAIDs and Cardiovascular Diseases: Role of Reactive Oxygen Species. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:536962. [PMID: 26457127 PMCID: PMC4592725 DOI: 10.1155/2015/536962] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 12/24/2022]
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are the most commonly used drugs worldwide. NSAIDs are used for a variety of conditions including pain, rheumatoid arthritis, and musculoskeletal disorders. The beneficial effects of NSAIDs in reducing or relieving pain are well established, and other benefits such as reducing inflammation and anticancer effects are also documented. The undesirable side effects of NSAIDs include ulcers, internal bleeding, kidney failure, and increased risk of heart attack and stroke. Some of these side effects may be due to the oxidative stress induced by NSAIDs in different tissues. NSAIDs have been shown to induce reactive oxygen species (ROS) in different cell types including cardiac and cardiovascular related cells. Increases in ROS result in increased levels of oxidized proteins which alters key intracellular signaling pathways. One of these key pathways is apoptosis which causes cell death when significantly activated. This review discusses the relationship between NSAIDs and cardiovascular diseases (CVD) and the role of NSAID-induced ROS in CVD.
Collapse
Affiliation(s)
- Rajeshwary Ghosh
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Azra Alajbegovic
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
| | - Aldrin V. Gomes
- Department of Neurobiology, Physiology, and Behavior, University of California, Davis, CA 95616, USA
- Department of Physiology and Membrane Biology, University of California, Davis, CA 95616, USA
| |
Collapse
|
14
|
Booth L, Roberts JL, Cruickshanks N, Tavallai S, Webb T, Samuel P, Conley A, Binion B, Young HF, Poklepovic A, Spiegel S, Dent P. PDE5 inhibitors enhance celecoxib killing in multiple tumor types. J Cell Physiol 2015; 230:1115-27. [PMID: 25303541 DOI: 10.1002/jcp.24843] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/02/2014] [Indexed: 12/20/2022]
Abstract
The present studies determined whether clinically relevant phosphodiesterase 5 (PDE5) inhibitors interacted with a clinically relevant NSAID, celecoxib, to kill tumor cells. Celecoxib and PDE5 inhibitors interacted in a greater than additive fashion to kill multiple tumor cell types. Celecoxib and sildenafil killed ex vivo primary human glioma cells as well as their associated activated microglia. Knock down of PDE5 recapitulated the effects of PDE5 inhibitor treatment; the nitric oxide synthase inhibitor L-NAME suppressed drug combination toxicity. The effects of celecoxib were COX2 independent. Over-expression of c-FLIP-s or knock down of CD95/FADD significantly reduced killing by the drug combination. CD95 activation was dependent on nitric oxide and ceramide signaling. CD95 signaling activated the JNK pathway and inhibition of JNK suppressed cell killing. The drug combination inactivated mTOR and increased the levels of autophagy and knock down of Beclin1 or ATG5 strongly suppressed killing by the drug combination. The drug combination caused an ER stress response; knock down of IRE1α/XBP1 enhanced killing whereas knock down of eIF2α/ATF4/CHOP suppressed killing. Sildenafil and celecoxib treatment suppressed the growth of mammary tumors in vivo. Collectively our data demonstrate that clinically achievable concentrations of celecoxib and sildenafil have the potential to be a new therapeutic approach for cancer.
Collapse
Affiliation(s)
- Laurence Booth
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Li D, Li L, Li P, Li Y, Chen X. Apoptosis of HeLa cells induced by a new targeting photosensitizer-based PDT via a mitochondrial pathway and ER stress. Onco Targets Ther 2015; 8:703-11. [PMID: 25897245 PMCID: PMC4396590 DOI: 10.2147/ott.s76370] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Photodynamic therapy (PDT) is emerging as a viable treatment for many cancers. To decrease the cutaneous photosensitivity induced by PDT, many attempts have been made to search for a targeting photosensitizer; however, few reports describe the molecular mechanism of PDT mediated by this type of targeting photosensitizer. The present study aimed to investigate the molecular mechanism of PDT induced by a new targeting photosensitizer (PS I), reported previously by us, on HeLa cells. Apoptosis is the primary mode of HeLa cell death in our system, and apoptosis occurs in a manner dependent on concentration, irradiation dose, and drug–light intervals. After endocytosis mediated by the folate receptor, PS I was primarily localized to the mitochondria and the endoplasmic reticulum (ER) of HeLa cells. PS I PDT resulted in rapid increases in intracellular reactive oxygen species (ROS) production and Ca2+ concentration, both of which reached a peak nearly simultaneously at 15 minutes, followed by the loss of mitochondrial membrane potential at 30 minutes, release of cytochrome c from mitochondria into the cytoplasm, downregulation of Bcl-2 expression, and upregulation of Bax expression. Meanwhile, activation of caspase-3, -9, and -12, as well as induction of C/EBP homologous protein (CHOP) and glucose-regulated protein (GRP78), in HeLa cells after PS I PDT was also detected. These results suggest that apoptosis of HeLa cells induced by PS I PDT is not only triggered by ROS but is also regulated by Ca2+ overload. Mitochondria and the ER serve as the subcellular targets of PS I PDT, the effective activation of which is responsible for PS I PDT-induced apoptosis in HeLa cells.
Collapse
Affiliation(s)
- Donghong Li
- State Key Laboratory of Trauma, Burn and Combined Injury, The Second Department of Research Institute of Surgery, Third Military Medical University, Chongqing, People's Republic of China
| | - Lei Li
- The First Department of Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Pengxi Li
- State Key Laboratory of Trauma, Burn and Combined Injury, The Second Department of Research Institute of Surgery, Third Military Medical University, Chongqing, People's Republic of China
| | - Yi Li
- Cancer Center, Daping Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Xiangyun Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, The Second Department of Research Institute of Surgery, Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
16
|
Song J, Wei Y, Chen Q, Xing D. Cyclooxygenase 2-mediated apoptotic and inflammatory responses in photodynamic therapy treated breast adenocarcinoma cells and xenografts. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2014; 134:27-36. [DOI: 10.1016/j.jphotobiol.2014.03.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 03/17/2014] [Accepted: 03/23/2014] [Indexed: 12/22/2022]
|