1
|
de Klerk DJ, de Keijzer MJ, Dias LM, Heemskerk J, de Haan LR, Kleijn TG, Franchi LP, Heger M. Strategies for Improving Photodynamic Therapy Through Pharmacological Modulation of the Immediate Early Stress Response. Methods Mol Biol 2022; 2451:405-480. [PMID: 35505025 DOI: 10.1007/978-1-0716-2099-1_20] [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: 06/14/2023]
Abstract
Photodynamic therapy (PDT) is a minimally to noninvasive treatment modality that has emerged as a promising alternative to conventional cancer treatments. PDT induces hyperoxidative stress and disrupts cellular homeostasis in photosensitized cancer cells, resulting in cell death and ultimately removal of the tumor. However, various survival pathways can be activated in sublethally afflicted cancer cells following PDT. The acute stress response is one of the known survival pathways in PDT, which is activated by reactive oxygen species and signals via ASK-1 (directly) or via TNFR (indirectly). The acute stress response can activate various other survival pathways that may entail antioxidant, pro-inflammatory, angiogenic, and proteotoxic stress responses that culminate in the cancer cell's ability to cope with redox stress and oxidative damage. This review provides an overview of the immediate early stress response in the context of PDT, mechanisms of activation by PDT, and molecular intervention strategies aimed at inhibiting survival signaling and improving PDT outcome.
Collapse
Affiliation(s)
- Daniel J de Klerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Mark J de Keijzer
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lionel M Dias
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Faculdade de Ciências da Saúde (FCS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Jordi Heemskerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
| | - Lianne R de Haan
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Tony G Kleijn
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Leonardo P Franchi
- Departamento de Bioquímica e Biologia Molecular, Instituto de Ciências Biológicas (ICB) 2, Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
- Faculty of Philosophy, Department of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group, Sciences, and Letters of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China.
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
2
|
Naderi M, Rahmani Cherati M, Mohammadian A, Baghery Bidhendy M, Ghiasvand S, Zare Marzouni H, Aryan H, Jangholi E, Javidi MA. Hypericin Induces Apoptosis in AGS Cell Line with No Significant Effect on Normal Cells. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 19:349-357. [PMID: 33680035 PMCID: PMC7758000 DOI: 10.22037/ijpr.2019.14904.12735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
It is of great importance to find an effective approach that not only eliminates gastric cancer cells but also do exhibits significant side effect to normal cells. Some studies have shown the effectiveness of hypericin against cancer cells. In this study, we evaluated the anti-cancer effect of Hypericin in the treatment of gastric cancer. In this study, the AGS cell line was exposed to different concentrations of hypericin for 24 and 48 h. Evaluation of cell death was done by MTT assay. The rate of apoptosis was measured by flow cytometry assay using Annexin V/ Propidium Iodide. The expression rate of Bcl2, p53 and Bax genes was evaluated by Real-time PCR test, and immunocytochemistry (ICC) analysis and western blotting was used for further evaluation of p53. MTT assay test showed that hyepricin induces 50% cell death in the concentration of 1 (µg/mL) and 0.5 (µg/mL) at 24 h and 48 h post-treatment, respectively, however no similar effect seen on fibroblast cells. Annexin/PI test revealed that cell apoptosis after exposure to hypericin for 24 h was 74%. Real-time PCR showed that expression level of Bax, p53 and Bax genes increases and Bcl2 gene decreases in AGS cell lines after treatment by hypericin. ICC analysis and western blotting for p53 confirmed these data. The results of this study indicated that hypericin has the potential to be introduced as an effective treatment for gastric cancer. Therefore, it seems that this substance has potential to be utilized as anti-cancer drug.
Collapse
Affiliation(s)
- Misagh Naderi
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mahtab Rahmani Cherati
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ali Mohammadian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Baghery Bidhendy
- Young Researchers and Elite Club, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran.,Clinical Research Development Center, Amir-almomenin Hospital, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Saeedeh Ghiasvand
- Departments of Biology, Faculty of Science, Malayer University, Malayer, Iran
| | - Hadi Zare Marzouni
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hoda Aryan
- Young Researchers and Elite Club, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran.,Medical Students' Scientific Association (MSSA), Tehran Medical Sciences Branch, Islamic Azad University Tehran, Iran
| | - Ehsan Jangholi
- Young Researchers and Elite Club, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran.,Clinical Research Development Center, Amir-almomenin Hospital, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Amin Javidi
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
3
|
Dong X, Zeng Y, Zhang Z, Fu J, You L, He Y, Hao Y, Gu Z, Yu Z, Qu C, Yin X, Ni J, Cruz LJ. Hypericin-mediated photodynamic therapy for the treatment of cancer: a review. J Pharm Pharmacol 2020; 73:425-436. [PMID: 33793828 DOI: 10.1093/jpp/rgaa018] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Hypericin is a polycyclic aromatic naphthodianthrone that occurs naturally. It is also an active ingredient in some species of the genus Hypericum. Emerging evidence suggests that hypericin has attracted great attention as a potential anticancer drug and exhibits remarkable antiproliferative effect upon irradiation on various tumour cells. This paper aims to summarise the anticancer effect and molecular mechanisms modulated by hypericin-medicated photodynamic therapy and its potential role in the cancer treatment. KEY FINDINGS Hypericin-medicated photodynamic therapy could inhibit the proliferation of various tumour cells including bladder, colon, breast, cervical, glioma, leukaemia, hepatic, melanoma, lymphoma and lung cancers. The effect is primarily mediated by p38 mitogen-activated protein kinase (MAPK), JNK, PI3K, CCAAT-enhancer-binding protein homologous protein (CHOP)/TRIB3/Akt/mTOR, TRAIL/TRAIL-receptor, c-Met and Ephrin-Eph, the mitochondria and extrinsic signalling pathways. Furthermore, hypericin-medicated photodynamic therapy in conjunction with chemotherapeutic agents or targeted therapies is more effective in inhibiting the growth of tumour cells. SUMMARY During the past few decades, the anticancer properties of photoactivated hypericin have been extensively investigated. Hypericin-medicated photodynamic therapy can modulate a variety of proteins and genes and exhibit a great potential to be used as a therapeutic agent for various types of cancer.
Collapse
Affiliation(s)
- Xiaoxv Dong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Leiden University Medical Center, Leiden, The Netherlands
| | - Yawen Zeng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhiqin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jing Fu
- Beijing Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Longtai You
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan He
- Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Leiden University Medical Center, Leiden, The Netherlands
| | - Yang Hao
- Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Leiden University Medical Center, Leiden, The Netherlands
| | - Zili Gu
- Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Leiden University Medical Center, Leiden, The Netherlands
| | - Zhenfeng Yu
- Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Leiden University Medical Center, Leiden, The Netherlands
| | - Changhai Qu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xingbin Yin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jian Ni
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Luis J Cruz
- Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
4
|
Chien CT, Chen YC, Liu YC, Liang SH, Lin HH, Lin CH. The antimicrobial photodynamic inactivation resistance of Candida albicans is modulated by the Hog1 pathway and the Cap1 transcription factor. Med Mycol 2019; 57:618-627. [PMID: 30289464 DOI: 10.1093/mmy/myy079] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 08/02/2018] [Accepted: 09/23/2018] [Indexed: 12/22/2022] Open
Abstract
Candida albicans is the most important fungal pathogen afflicting humans, particularly immunocompromised patients. However, currently available antifungal drugs are limited and ineffective against drug-resistant strains. The development of new drugs or alternative therapeutic approaches to control fungal infections is urgent and necessary. Photodynamic inactivation (PDI) is a new promising therapy for eradicating microorganism infections through combining visible light, photosensitizers, and oxygen to generate reactive oxygen species (ROS). Although cytoprotective responses induced by photodynamic therapy (PDT) have been well studied in cancer cells, the mechanisms by which C. albicans responds to PDI are largely unknown. In this study, we first demonstrated that PDI induces C. albicans Hog1p activation. Deletion of any of the SSK2, PBS2, and HOG1 genes significantly decreased the survival rate after photochemical reactions, indicating that the Hog1 SAPK pathway is required for tolerance to PDI. Furthermore, the basic leucine zipper transcription factor Cap1 that regulates several downstream antioxidant genes was highly expressed during the response to PDI, and loss of CAP1 also resulted in decreased C. albicans survival rates. This study demonstrates the importance of the Hog1 SAPK and the Cap1 transcription factor, which regulates in resistance to PDI-mediated oxidative stress in C. albicans. Understanding the mechanisms by which C. albicans responds to PDI and consequently scavenges ROS will be very useful for the further development of therapeutics to control fungal infectious diseases, particularly those of the skin and mucosal infections.
Collapse
Affiliation(s)
- Chih-Ting Chien
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yu-Chia Chen
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yun-Chun Liu
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | | | - Hsien-Hen Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Ching-Hsuan Lin
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Wu CX, Feng YH, Yang L, Zhan ZL, Xu XH, Hu XY, Zhu ZH, Zhou GP. Electroacupuncture exerts neuroprotective effects on ischemia/reperfusion injury in JNK knockout mice: the underlying mechanism. Neural Regen Res 2018; 13:1594-1601. [PMID: 30127120 PMCID: PMC6126120 DOI: 10.4103/1673-5374.235294] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Simple regulation of c-Jun N-terminal kinase (JNK) or p38 mitogen-activated protein kinase (MAPK) pathways is not enough to trigger cell apoptosis. However, activation of the stress activated pathway (JNK/p38 MAPK) together with inhibition of the growth factor activated extracellular signal-regulated kinase (ERK) pathway can promote cell apoptosis. We hypothesized that inhibition of the JNK or p38 pro-apoptotic pathway and activating the ERK pathway could be the mechanism of anti-apoptosis following cerebral ischemia/reperfusion injury. To investigate the mechanism of the protective effect of electroacupuncture on cerebral ischemia/reperfusion injury in JNK knockout mice, mouse models of cerebral ischemia/reperfusion injury were established by Longa’s method. Electroacupuncture was conducted at acupoints Chize (LU5), Hegu (LI4), Sanyinjiao (SP6) and Zusanli (ST36) 1.5 hours after ischemia/reperfusion injury for 20 minutes, once a day. The neurological function was evaluated using neurological deficit scores. The expression of phospho-extracellular signal-regulated kinase (p-ERK) and phospho-p38 (p-p38) in JNK knockout mice was detected using double-labeling immunofluorescence and western blot assay. The mRNA expression of ERK and p38 was measured by quantitative real-time polymerase chain reaction. Electroacupuncture improved neurological function, increased the immunoreactivity and relative expression of p-ERK and reduced that of p-p38 in the cerebral cortex and hippocampus on the injured side. Electroacupuncture increased mRNA expression of ERK, but decreased that of p38 in the cerebral cortex and hippocampus on the injured side. In conclusion, electroacupuncture upregulated the protective ERK pathway and inhibited the pro-apoptotic p38 pathway, thereby exerting a neuroprotective effect and improving the neurological function in JNK knockout mice.
Collapse
Affiliation(s)
- Chun-Xiao Wu
- School of Traditional Chinese Medicine, Southern Medical University; Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Yi-Hui Feng
- Central Hospital of Qingdao, Qingdao, Shandong Province, China
| | - Lu Yang
- School of Traditional Chinese Medicine, Southern Medical University; Traditional Chinese Medicine-Integrated Hospital, the Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhu-Lian Zhan
- Traditional Chinese Medicine-Integrated Hospital, the Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiu-Hong Xu
- Traditional Chinese Medicine-Integrated Hospital, the Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiao-Ying Hu
- Traditional Chinese Medicine-Integrated Hospital, the Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhi-Hua Zhu
- Ruikang Hospital Affiliated to Guangxi College of Traditional Chinese Medicine, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Guo-Ping Zhou
- School of Traditional Chinese Medicine, Southern Medical University; Traditional Chinese Medicine-Integrated Hospital, the Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
6
|
Li A, Shi D, Xu B, Wang J, Tang YL, Xiao W, Shen G, Deng W, Zhao C. S100A6 promotes cell proliferation in human nasopharyngeal carcinoma via the p38/MAPK signaling pathway. Mol Carcinog 2016; 56:972-984. [PMID: 27596819 DOI: 10.1002/mc.22563] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 08/25/2016] [Accepted: 09/04/2016] [Indexed: 01/15/2023]
Abstract
An elevated level of S100A6 is associated with poor outcomes of many tumor types, but, how S100A6 contributes to nasopharyngeal carcinoma (NPC) progression remains unknown. Here, we investigated the expression and prognostic significance of S100A6 in NPC and explored the molecular mechanisms under-lying the role of S100A6 in NPC development. The results showed that S100A6 was markedly up-regulated in NPC tissues and cell lines compared to paired peritumoral normal tissues and a normal nasopharyngeal epithelial cell line, respectively. In tissues from 92 NPC patients, high S100A6 expression was associated with advanced N stage, locoregional failure and disease progression and was predictive of poor locoregional recurrence-free survival (LRRFS, P = 0.001) and progression-free survival (PFS, P = 0.001). Multivariate analysis showed that S100A6 is an independent prognostic factor for LRRFS and PFS. Silencing S100A6 using siRNA or shRNA significantly suppressed NPC cell proliferation, colony formation and p38/mitogen-activated protein kinase (MAPK) activity in vitro and inhibited tumor growth in a xenograft mouse model of NPC. In contrast, overexpressing S100A6 via plasmid transfection resulted in increased NPC cell proliferation and p38/MAPK activation. S100A6-induced proliferation was abolished by a p38 inhibitor. In summary, S100A6 may be a new prognostic marker of NPC and may promote NPC development via the activation of p38/MAPK signaling pathways. These findings suggest S100A6/p38/MAPK signaling as a potential therapeutic target for NPC. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Anchuan Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China.,Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Dingbo Shi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Benhua Xu
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jingshu Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan-Lai Tang
- Department of Pediatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - WeiWei Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guanzhu Shen
- Department of Radiation Oncology, Cancer Center of Guangzhou Medical University, Guangzhou, China
| | - Wuguo Deng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chong Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
7
|
Broekgaarden M, Weijer R, van Gulik TM, Hamblin MR, Heger M. Tumor cell survival pathways activated by photodynamic therapy: a molecular basis for pharmacological inhibition strategies. Cancer Metastasis Rev 2015; 34:643-90. [PMID: 26516076 PMCID: PMC4661210 DOI: 10.1007/s10555-015-9588-7] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Photodynamic therapy (PDT) has emerged as a promising alternative to conventional cancer therapies such as surgery, chemotherapy, and radiotherapy. PDT comprises the administration of a photosensitizer, its accumulation in tumor tissue, and subsequent irradiation of the photosensitizer-loaded tumor, leading to the localized photoproduction of reactive oxygen species (ROS). The resulting oxidative damage ultimately culminates in tumor cell death, vascular shutdown, induction of an antitumor immune response, and the consequent destruction of the tumor. However, the ROS produced by PDT also triggers a stress response that, as part of a cell survival mechanism, helps cancer cells to cope with the PDT-induced oxidative stress and cell damage. These survival pathways are mediated by the transcription factors activator protein 1 (AP-1), nuclear factor E2-related factor 2 (NRF2), hypoxia-inducible factor 1 (HIF-1), nuclear factor κB (NF-κB), and those that mediate the proteotoxic stress response. The survival pathways are believed to render some types of cancer recalcitrant to PDT and alter the tumor microenvironment in favor of tumor survival. In this review, the molecular mechanisms are elucidated that occur post-PDT to mediate cancer cell survival, on the basis of which pharmacological interventions are proposed. Specifically, pharmaceutical inhibitors of the molecular regulators of each survival pathway are addressed. The ultimate aim is to facilitate the development of adjuvant intervention strategies to improve PDT efficacy in recalcitrant solid tumors.
Collapse
Affiliation(s)
- Mans Broekgaarden
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Ruud Weijer
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Thomas M van Gulik
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA, USA
| | - Michal Heger
- Department of Experimental Surgery, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
8
|
Tsai YJ, Tsai T, Peng PC, Li PT, Chen CT. Histone acetyltransferase p300 is induced by p38MAPK after photodynamic therapy: the therapeutic response is increased by the p300HAT inhibitor anacardic acid. Free Radic Biol Med 2015; 86:118-32. [PMID: 26001729 DOI: 10.1016/j.freeradbiomed.2015.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 05/07/2015] [Accepted: 05/08/2015] [Indexed: 12/11/2022]
Abstract
Oxidative stress mediated by photodynamic therapy (PDT) mediates the tumoricidal effect, but has also been shown to induce the expression of prosurvival molecules, such as cyclooxygenase-2 (COX-2), which is involved in tumor recurrences after PDT. However, the molecular mechanism is still not fully understood. In this study, we found that activated p38MAPK could significantly up-regulate the activity and expression of histone acetyltransferase p300 (p300HAT) in A375 and C26 cells treated with ALA-and chlorin e6 (Ce6)-mediated photodynamic treatment. A colony-formation assay showed that PDT-induced cytotoxicity was dramatically elevated in the presence of the p300HAT inhibitor anacardic acid (AA). Further studies showed that increased p300HAT acetylates histone H3 and NF-κB p65 subunit to up-regulate the COX-2 expression, which was reduced by AA or p300HAT shRNA. Using chromatin immunoprecipitation analysis, we found that the augmented acetylation of histone H3 and NF-κB increases their binding to the COX-2 promoter region. These in vitro findings were further verified in mice bearing murine C26 and human A375 tumors treated with liposomal Ce6 mediated PDT. Meanwhile, the combination of PDT and AA resulted in greater tumor regression in BALB/c mice bearing C26 tumors, compared with PDT only or combined with COX-2 inhibitor. Finally, we demonstrated that suppression of the PDT-induced p300HAT activity also resulted in the decreased expression of survivin, restoring caspase-3 activity and sensitizing PDT-treated cells from autophagy to apoptosis due to the Becline-1 cleavage. This study demonstrates for the first time the molecular mechanisms involved in histone modification induced by PDT-mediated oxidative stress, suggesting that HAT inhibitors may provide a novel therapeutic approach for improving PDT response.
Collapse
Affiliation(s)
- Yi-Jane Tsai
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Tsuimin Tsai
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Po-Chun Peng
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Pei-Tzu Li
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Chin-Tin Chen
- Department of Biochemical Science and Technology, National Taiwan University, Taipei 10617, Taiwan.
| |
Collapse
|
9
|
Chan KC, Ting CM, Chan PS, Lo MC, Lo KW, Curry JE, Smyth T, Lee AWM, Ng WT, Tsao GSW, Wong RNS, Lung ML, Mak NK. A novel Hsp90 inhibitor AT13387 induces senescence in EBV-positive nasopharyngeal carcinoma cells and suppresses tumor formation. Mol Cancer 2013; 12:128. [PMID: 24156782 PMCID: PMC3834878 DOI: 10.1186/1476-4598-12-128] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 10/02/2013] [Indexed: 02/06/2023] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is an epithelial malignancy strongly associated with Epstein-Barr virus (EBV). AT13387 is a novel heat shock protein 90 (Hsp90) inhibitor, which inhibits the chaperone function of Hsp90 and reduces expression of Hsp90-dependent client oncoproteins. This study aimed to evaluate both the in vitro and in vivo antitumor effects of AT13387 in the EBV-positive NPC cell line C666-1. Results Our results showed that AT13387 inhibited C666-1 cell growth and induced cellular senescence with the downregulation of multiple Hsp90 client oncoproteins EGFR, AKT, CDK4, and restored the protein expression of negative cell cycle regulator p27. We also studied the ability of AT13387 to restore p27 expression by downregulation of AKT and the p27 ubiquitin mediator, Skp2, using AKT inhibitor and Skp2 siRNA. In the functional study, AT13387 inhibited cell migration with downregulation of a cell migration regulator, HDAC6, and increased the acetylation and stabilization of α-tubulin. We also examined the effect of AT13387 on putative cancer stem cells (CSC) by 3-D tumor sphere formation assay. AT13387 effectively reduced both the number and size of C666-1 tumor spheres with decreased expression of NPC CSC-like markers CD44 and SOX2. In the in vivo study, AT13387 significantly suppressed tumor formation in C666-1 NPC xenografts. Conclusion AT13387 suppressed cell growth, cell migration, tumor sphere formation and induced cellular senescence on EBV-positive NPC cell line C666-1. Also, the antitumor effect of AT13387 was demonstrated in an in vivo model. This study provided experimental evidence for the preclinical value of using AT13387 as an effective antitumor agent in treatment of NPC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Nai Ki Mak
- Department of Biology, Hong Kong Baptist University, Hong Kong, P,R, China.
| |
Collapse
|
10
|
Keskin T, Isgor BS, Isgor YG, Yukruk F. Evaluation of Perylenediimide Derivatives for Potential Therapeutic Benefits on Cancer Chemotherapy. Chem Biol Drug Des 2012; 80:675-81. [DOI: 10.1111/cbdd.12004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
11
|
Hu ZY, Wang J, Cheng G, Zhu XF, Huang P, Yang D, Zeng YX. Apogossypolone targets mitochondria and light enhances its anticancer activity by stimulating generation of singlet oxygen and reactive oxygen species. CHINESE JOURNAL OF CANCER 2012; 30:41-53. [PMID: 21192843 PMCID: PMC4012262 DOI: 10.5732/cjc.010.10295] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Apogossypolone (ApoG2), a novel derivative of gossypol, has been shown to be a potent inhibitor of antiapoptotic Bcl-2 family proteins and to have antitumor activity in multiple types of cancer cells. Recent reports suggest that gossypol stimulates the generation of cellular reactive oxygen species (ROS) in leukemia and colorectal carcinoma cells; however, gossypol-mediated cell death in leukemia cells was reported to be ROS-independent. This study was conducted to clarify the effect of ApoG2-induced ROS on mitochondria and cell viability, and to further evaluate its utility as a treatment for nasopharyngeal carcinoma (NPC). We tested the photocytotoxicity of ApoG2 to the poorly differentiated NPC cell line CNE-2 using the ROS-generating TL/10 illumination system. The rapid ApoG2-induced cell death was partially reversed by the antioxidant N-acetyl-L-cysteine (NAC), but the ApoG2-induced reduction of mitochondrial membrane potential (MMP) was not reversed by NAC. In the presence of TL/10 illumination, ApoG2 generated massive amounts of singlet oxygen and was more effective in inhibiting cell growth than in the absence of illumination. We also determined the influence of light on the anti-proliferative activity of ApoG2 using a CNE-2–xenograft mouse model. ApoG2 under TL/10 illumination healed tumor wounds and suppressed tumor growth more effectively than ApoG2 treatment alone. These results indicate that the ApoG2-induced CNE-2 cell death is partly ROS-dependent. ApoG2 may be used with photodynamic therapy (PDT) to treat NPC.
Collapse
Affiliation(s)
- Zhe-Yu Hu
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, PR China
| | | | | | | | | | | | | |
Collapse
|
12
|
Zhao Z, Chan PS, Li H, Wong KL, Wong RNS, Mak NK, Zhang J, Tam HL, Wong WY, Kwong DWJ, Wong WK. Highly Selective Mitochondria-Targeting Amphiphilic Silicon(IV) Phthalocyanines with Axially Ligated Rhodamine B for Photodynamic Therapy. Inorg Chem 2011; 51:812-21. [DOI: 10.1021/ic201178e] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Zhixin Zhao
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
| | - Pui-Shan Chan
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
| | - Hongguang Li
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
| | - Ka-Leung Wong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
| | - Ricky Ngok Shun Wong
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
| | - Nai-Ki Mak
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
| | - Jie Zhang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
| | - Hoi-Lam Tam
- Department of Physics, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
| | - Wai-Yeung Wong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
- Institute of Molecular Functional Materials, Areas of Excellence Schemes, University
Grants Committee, Hong Kong
| | - Daniel W. J. Kwong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
| | - Wai-Kwok Wong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong
SAR
- Institute of Molecular Functional Materials, Areas of Excellence Schemes, University
Grants Committee, Hong Kong
| |
Collapse
|
13
|
Hypericins as potential leads for new therapeutics. Int J Mol Sci 2010; 11:562-94. [PMID: 20386655 PMCID: PMC2852855 DOI: 10.3390/ijms11020562] [Citation(s) in RCA: 175] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2009] [Revised: 01/27/2010] [Accepted: 01/28/2010] [Indexed: 12/22/2022] Open
Abstract
70 years have passed since the first isolation of the naphthodianthrones hypericin and pseudohypericin from Hypericum perforatum L. Today, they continue to be one of the most promising group of polyphenols, as they fascinate with their physical, chemical and important biological properties which derive from their unique chemical structure. Hypericins and their derivatives have been extensively studied mainly for their antitumor, antiviral and antidepressant properties. Notably, hypericin is one of the most potent naturally occurring photodynamic agents. It is able to generate the superoxide anion and a high quantum yield of singlet oxygen that are considered to be primarily responsible for its biological effects. The prooxidant photodynamic properties of hypericin have been exploited for the photodynamic therapy of cancer (PDT), as hypericin, in combination with light, very effectively induces apoptosis and/or necrosis of cancer cells. The mechanism by which these activities are expressed continues to be a main topic of discussion, but according to scientific data, different modes of action (generation of ROS & singlet oxygen species, antiangiogenesis, immune responces) and multiple molecular pathways (intrinsic/extrinsic apoptotic pathway, ERK inhibition) possibly interrelating are implicated. The aim of this review is to analyse the most recent advances (from 2005 and thereof) in the chemistry and biological activities (in vitro and in vivo) of the pure naphthodianthrones, hypericin and pseudohypericin from H. perforatum. Extracts from H. perforatum were not considered, nor pharmakokinetic or clinical data. Computerised literature searches were performed using the Medline (PubMed), ChemSciFinder and Scirus Library databases. No language restrictions were imposed.
Collapse
|