1
|
Zhang M, Liu X, Ran X, Gao R, Sun J, Zhuang K, You Z, Zhang Z, Ran Y. Hypocrellin A-mediated photodynamic antibacterial activity against Cutibacterium acnes: An in vitro study. Photodiagnosis Photodyn Ther 2025; 51:104467. [PMID: 39798778 DOI: 10.1016/j.pdpdt.2024.104467] [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: 12/01/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025]
Abstract
Skin dysbiosis caused by Cutibacterium acnes contributes greatly to the complex pathogenesis of acne, and antimicrobial photodynamic therapy (PDT) has emerged as a promising treatment option for acne treatment. Hypocrellin, a photosensitizer extracted from a traditional Chinese medicinal fungus, has showed effective antimicrobial activity. This study aimed to evaluate the antibacterial ability of hypocrellin mediated PDT against Cutibacterium acnes. Using modified broth dilution method and morphological observation, the antibacterial effect was tested under a series of experimental conditions. The results showed that hypocrellin initiates type II photodynamic reactions by inducing amount of reactive oxygen species, particularly singlet oxygen. Within a certain concentration range, hypocrellin effectively maintained the antibacterial efficacy with minimal damage to keratinocyte cells. These results provide new insights into the use of PDT for acne treatment.
Collapse
Affiliation(s)
- Muqiu Zhang
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyao Liu
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Pathogen Research, West China Hospital, Sichuan University, Chengdu, China.
| | - Xin Ran
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Runyan Gao
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jitong Sun
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Kaiwen Zhuang
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zimeng You
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Zhi Zhang
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yuping Ran
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, China; Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
2
|
Gugu Nkosi PW, Chandran R, Abrahamse H. Hypocrellin: A Natural Photosensitizer and Nano-Formulation for Enhanced Molecular Targeting of PDT of Melanoma. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1997. [PMID: 39568119 PMCID: PMC11579242 DOI: 10.1002/wnan.1997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/29/2024] [Accepted: 09/10/2024] [Indexed: 11/22/2024]
Abstract
Nano-formulation has generated attention in the battle against cancer, because of its great flexibility, reduced adverse side effects, and accuracy in delivering drugs to target tissues dependent on the size and surface characteristics of the disease. The field of photodynamic treatment has advanced significantly in the past years. Photodynamic techniques that use nano-formulations have surfaced to further the field of nanotechnology in medicine, especially in cancer treatment. The pharmaceutical industry is seeing a growing trend toward enhanced drug formulation using nano-formulations such as liposomes, polymeric nanoparticles, dendrimers, nano-emulsions, and micelles. Natural extracts have also shown adverse effects when employed as photosensitizers in cancer therapy because they are cytotoxic when activated by light. Still, natural photosensitizers are a big part of cancer treatment. However, some shortcomings can be minimized by combining nano-formulations with these natural photosensitizers. The synergistic improvement in medication delivery that maintains or increases the mechanism of cell death in malignant cells has also been demonstrated by the combination of photodynamic therapy with nano-formulations and natural photosensitizers. Lastly, this review assesses the feasibility and potential of a photodynamic therapy system based on nano-formulations and natural photosensitizers in clinical treatment applications and briefly discusses the removal of toxic compounds associated with nano-formulations within cells.
Collapse
Affiliation(s)
| | - Rahul Chandran
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgDoornfonteinSouth Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health SciencesUniversity of JohannesburgDoornfonteinSouth Africa
| |
Collapse
|
3
|
Boo YC. Insights into How Plant-Derived Extracts and Compounds Can Help in the Prevention and Treatment of Keloid Disease: Established and Emerging Therapeutic Targets. Int J Mol Sci 2024; 25:1235. [PMID: 38279232 PMCID: PMC10816582 DOI: 10.3390/ijms25021235] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Keloid is a disease in which fibroblasts abnormally proliferate and synthesize excessive amounts of extracellular matrix, including collagen and fibronectin, during the healing process of skin wounds, causing larger scars that exceed the boundaries of the original wound. Currently, surgical excision, cryotherapy, radiation, laser treatment, photodynamic therapy, pressure therapy, silicone gel sheeting, and pharmacotherapy are used alone or in combinations to treat this disease, but the outcomes are usually unsatisfactory. The purpose of this review is to examine whether natural products can help treat keloid disease. I introduce well-established therapeutic targets for this disease and various other emerging therapeutic targets that have been proposed based on the phenotypic difference between keloid-derived fibroblasts (KFs) and normal epidermal fibroblasts (NFs). We then present recent studies on the biological effects of various plant-derived extracts and compounds on KFs and NFs. Associated ex vivo, in vivo, and clinical studies are also presented. Finally, we discuss the mechanisms of action of the plant-derived extracts and compounds, the pros and cons, and the future tasks for natural product-based therapy for keloid disease, as compared with existing other therapies. Extracts of Astragalus membranaceus, Salvia miltiorrhiza, Aneilema keisak, Galla Chinensis, Lycium chinense, Physalis angulate, Allium sepa, and Camellia sinensis appear to modulate cell proliferation, migration, and/or extracellular matrix (ECM) production in KFs, supporting their therapeutic potential. Various phenolic compounds, terpenoids, alkaloids, and other plant-derived compounds could modulate different cell signaling pathways associated with the pathogenesis of keloids. For now, many studies are limited to in vitro experiments; additional research and development are needed to proceed to clinical trials. Many emerging therapeutic targets could accelerate the discovery of plant-derived substances for the prevention and treatment of keloid disease. I hope that this review will bridge past, present, and future research on this subject and provide insight into new therapeutic targets and pharmaceuticals, aiming for effective keloid treatment.
Collapse
Affiliation(s)
- Yong Chool Boo
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea;
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, The Graduate School, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| |
Collapse
|
4
|
Abou Shousha S, Osman EM, Baheeg S, Shahine Y. Anti-IL-8 monoclonal antibodies inhibits the autophagic activity and cancer stem cells maintenance within breast cancer tumor microenvironment. Breast Dis 2024; 43:37-49. [PMID: 38552109 PMCID: PMC10977415 DOI: 10.3233/bd-230052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
BACKGROUND Breast cancer tumor microenvironment (TME) is a promising target for immunotherapy. Autophagy, and cancer stem cells (CSCs) maintenance are essential processes involved in tumorigenesis, tumor survival, invasion, and treatment resistance. Overexpression of angiogenic chemokine interleukin-8 (IL-8) in breast cancer TME is associated with oncogenic signaling pathways, increased tumor growth, metastasis, and poor prognosis. OBJECTIVE Thus, we aimed to investigate the possible anti-tumor effect of neutralizing antibodies against IL-8 by evaluating its efficacy on autophagic activity and breast CSC maintenance. METHODS IL-8 monoclonal antibody supplemented tumor tissue culture systems from 15 females undergoing mastectomy were used to evaluate the expression of LC3B as a specific biomarker of autophagy and CD44, CD24 as cell surface markers of breast CSCs using immunofluorescence technique. RESULTS Our results revealed that anti-IL-8 mAb significantly decreased the level of LC3B in the cultured tumor tissues compared to its non-significant decrease in the normal breast tissues.Anti-IL-8 mAb also significantly decreased the CD44 expression in either breast tumors or normal cultured tissues. While it caused a non-significant decrease in CD24 expression in cultured breast tumor tissue and a significant decrease in its expression in the corresponding normal ones. CONCLUSIONS Anti-IL-8 monoclonal antibody exhibits promising immunotherapeutic properties through targeting both autophagy and CSCs maintenance within breast cancer TME.
Collapse
Affiliation(s)
- Seham Abou Shousha
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Eman M. Osman
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Suzan Baheeg
- Immunology and Allergy Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Yasmine Shahine
- Faculty of Pharmacy, Department of Microbiology & Immunology, Pharos University in Alexandria, Alexandria, Egypt
| |
Collapse
|
5
|
Gulia S, Chandra P, Das A. The Prognosis of Cancer Depends on the Interplay of Autophagy, Apoptosis, and Anoikis within the Tumor Microenvironment. Cell Biochem Biophys 2023; 81:621-658. [PMID: 37787970 DOI: 10.1007/s12013-023-01179-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2023] [Indexed: 10/04/2023]
Abstract
Within the tumor microenvironment, the fight between the immune system and cancer influences tumor transformation. Metastasis formation is an important stage in the progression of cancer. This process is aided by cellular detachment and resistance to anoikis, which are achieved by altering intercellular signaling. Autophagy, specifically pro-survival autophagy, aids cancer cells in developing treatment resistance. Numerous studies have shown that autophagy promotes tumor growth and resistance to anoikis. To regulate protective autophagy, cancer-related genes phosphorylate both pro- and anti-apoptotic proteins. Apoptosis, a type of controlled cell death, eliminates damaged or unwanted cells. Anoikis is a type of programmed cell death in which cells lose contact with the extracellular matrix. The dysregulation of these cellular pathways promotes tumor growth and spread. Apoptosis, anoikis, and autophagy interact meticulously and differently depending on the cellular circumstances. For instance, autophagy can protect cancer cells from apoptosis by removing cellular components that are damaged and might otherwise trigger apoptotic pathways. Similarly, anoikis dysregulation can trigger autophagy by causing cellular harm and metabolic stress. In order to prevent or treat metastatic disease, specifically, targeting these cellular mechanisms may present a promising prospect for cancer therapy. This review discourses the state of our understanding of the molecular and cellular mechanisms underlying tumor transformation and the establishment of metastatic tumors. To enhance the prognosis for cancer, we highlight and discuss potential therapeutic approaches that target these processes and genes involved in them.
Collapse
Affiliation(s)
- Shweta Gulia
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi, 110042, India
| | - Prakash Chandra
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi, 110042, India
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Main Bawana Road, Delhi, 110042, India.
| |
Collapse
|
6
|
Bao Z, Xie Y, Xu C, Zhang Z, Zhu D. Biotechnological production and potential applications of hypocrellins. Appl Microbiol Biotechnol 2023; 107:6421-6438. [PMID: 37695342 DOI: 10.1007/s00253-023-12727-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/01/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023]
Abstract
Hypocrellins (HYPs), a kind of natural perylenequinones (PQs) with an oxidized pentacyclic core, are important natural compounds initially extracted from the stromata of Hypocrella bambusae and Shiraia bambusicola. They have been widely concerned for their use as anti-microbial, anti-cancers, and anti-viral photodynamic therapy agents in recent years. Considering the restrictions of natural stromal resources, submerged fermentation with Shiraia spp. has been viewed as a promising alternative biotechnology for HYP production, and great efforts have been made to improve HYP production over the past decade. This article reviews recent publications about the mycelium fermentation production of HYPs, and their bioactivities and potential applications, and especially summarizes the progresses toward manipulation of fermentation conditions. Also, their chemical structure and analytic methods are outlined. Herein, it is worth mentioning that the gene arrangement in HYP gene cluster is revised; previous unknown genes in HYP and CTB gene clusters with correct function annotation are deciphered; the homologous sequences of HYP, CTB, and elc are systematically aligned, and especially the biosynthetic pathway of HYPs is full-scale proposed. KEY POINTS: • The mycelial fermentation process and metabolic regulation of hypocrellins are reviewed. • The bioactivities and potential applications of hypocrellins are summarized. • The biosynthesis pathway and regulatory mechanisms of hypocrellins are outlined.
Collapse
Affiliation(s)
- Zhuanying Bao
- Key Laboratory of Protection and Utilization of Subtropic Plant Resources of Jiangxi Province, Jiangxi Normal University, Nanchang, 330022, China
| | - Yunchang Xie
- Key Laboratory of Protection and Utilization of Subtropic Plant Resources of Jiangxi Province, Jiangxi Normal University, Nanchang, 330022, China
- Key Laboratory of Bioprocess Engineering of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China
| | - Chenglong Xu
- Key Laboratory of Protection and Utilization of Subtropic Plant Resources of Jiangxi Province, Jiangxi Normal University, Nanchang, 330022, China
| | - Zhibin Zhang
- Key Laboratory of Protection and Utilization of Subtropic Plant Resources of Jiangxi Province, Jiangxi Normal University, Nanchang, 330022, China
| | - Du Zhu
- Key Laboratory of Protection and Utilization of Subtropic Plant Resources of Jiangxi Province, Jiangxi Normal University, Nanchang, 330022, China.
- Key Laboratory of Bioprocess Engineering of Jiangxi Province, College of Life Sciences, Jiangxi Science and Technology Normal University, Nanchang, 330013, China.
| |
Collapse
|
7
|
Deng H, Liang X, Liu J, Zheng X, Fan TP, Cai Y. Advances and perspectives on perylenequinone biosynthesis. Front Microbiol 2022; 13:1070110. [PMID: 36605511 PMCID: PMC9808054 DOI: 10.3389/fmicb.2022.1070110] [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: 10/14/2022] [Accepted: 11/21/2022] [Indexed: 12/24/2022] Open
Abstract
Under illumination, the fungal secondary metabolites, perylenequinones (PQs) react with molecular oxygen to generate reactive oxygen species (ROS), which, in excess can damage cellular macromolecules and trigger apoptosis. Based on this property, PQs have been widely used as photosensitizers and applied in pharmaceuticals, which has stimulated research into the discovery of new PQs and the elucidation of their biosynthetic pathways. The PQs-associated literature covering from April 1967 to September 2022 is reviewed in three sections: (1) the sources, structural diversity, and biological activities of microbial PQs; (2) elucidation of PQ biosynthetic pathways, associated genes, and mechanisms of regulation; and (3) advances in pathway engineering and future potential strategies to modify cellular metabolism and improve PQ production.
Collapse
Affiliation(s)
- Huaxiang Deng
- Center for Synthetic Biochemistry, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China,The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China,*Correspondence: Huaxiang Deng,
| | - Xinxin Liang
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jinbin Liu
- School of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, Jiangsu, China
| | - Xiaohui Zheng
- College of Life Sciences, Northwest University, Xi’an, Shanxi, China
| | - Tai-Ping Fan
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
| | - Yujie Cai
- The Key Laboratory of Industrial Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China,Yujie Cai,
| |
Collapse
|
8
|
Carigga Gutierrez NM, Pujol-Solé N, Arifi Q, Coll JL, le Clainche T, Broekgaarden M. Increasing cancer permeability by photodynamic priming: from microenvironment to mechanotransduction signaling. Cancer Metastasis Rev 2022; 41:899-934. [PMID: 36155874 DOI: 10.1007/s10555-022-10064-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/06/2022] [Indexed: 01/25/2023]
Abstract
The dense cancer microenvironment is a significant barrier that limits the penetration of anticancer agents, thereby restraining the efficacy of molecular and nanoscale cancer therapeutics. Developing new strategies to enhance the permeability of cancer tissues is of major interest to overcome treatment resistance. Nonetheless, early strategies based on small molecule inhibitors or matrix-degrading enzymes have led to disappointing clinical outcomes by causing increased chemotherapy toxicity and promoting disease progression. In recent years, photodynamic therapy (PDT) has emerged as a novel approach to increase the permeability of cancer tissues. By producing excessive amounts of reactive oxygen species selectively in the cancer microenvironment, PDT increases the accumulation, penetration depth, and efficacy of chemotherapeutics. Importantly, the increased cancer permeability has not been associated to increased metastasis formation. In this review, we provide novel insights into the mechanisms by which this effect, called photodynamic priming, can increase cancer permeability without promoting cell migration and dissemination. This review demonstrates that PDT oxidizes and degrades extracellular matrix proteins, reduces the capacity of cancer cells to adhere to the altered matrix, and interferes with mechanotransduction pathways that promote cancer cell migration and differentiation. Significant knowledge gaps are identified regarding the involvement of critical signaling pathways, and to which extent these events are influenced by the complicated PDT dosimetry. Addressing these knowledge gaps will be vital to further develop PDT as an adjuvant approach to improve cancer permeability, demonstrate the safety and efficacy of this priming approach, and render more cancer patients eligible to receive life-extending treatments.
Collapse
Affiliation(s)
| | - Núria Pujol-Solé
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Qendresa Arifi
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Jean-Luc Coll
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Tristan le Clainche
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France.
| | - Mans Broekgaarden
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France.
| |
Collapse
|
9
|
Xiao Q, Wang L, Zhang J, Zhong X, Guo Z, Yu J, Ma Y, Wu H. Activation of Wnt/β-Catenin Signaling Involves 660 nm Laser Radiation on Epithelium and Modulates Lipid Metabolism. Biomolecules 2022; 12:1389. [PMID: 36291598 PMCID: PMC9599573 DOI: 10.3390/biom12101389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/10/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Research has proven that light treatment, specifically red light radiation, can provide more clinical benefits to human health. Our investigation was firstly conducted to characterize the tissue morphology of mouse breast post 660 nm laser radiation with low power and long-term exposure. RNA sequencing results revealed that light exposure with a higher intervention dosage could cause a number of differentially expressed genes compared with a low intervention dosage. Gene ontology analysis, protein-protein interaction network analysis, and gene set enrichment analysis results suggested that 660 nm light exposure can activate more transcription-related pathways in HC11 breast epithelial cells, and these pathways may involve modulating critical gene expression. To consider the critical role of the Wnt/T-catenin pathway in light-induced modulation, we hypothesized that this pathway might play a major role in response to 660 nm light exposure. To validate our hypothesis, we conducted qRT-PCR, immunofluorescence staining, and Western blot assays, and relative results corroborated that laser radiation could promote expression levels of β-catenin and relative phosphorylation. Significant changes in metabolites and pathway analysis revealed that 660 nm laser could affect nucleotide metabolism by regulating purine metabolism. These findings suggest that the Wnt/β-catenin pathway may be the major sensor for 660 nm laser radiation, and it may be helpful to rescue drawbacks or side effects of 660 nm light exposure through relative interventional agents.
Collapse
Affiliation(s)
- Qiyang Xiao
- School of Artificial Intelligence, Henan University, Zhengzhou 450046, China
| | - Lijing Wang
- School of Life Sciences, Henan University, Kaifeng 475000, China
| | - Juling Zhang
- Center for Faculty Development, South China Normal University, Guangzhou 510631, China
| | - Xinyu Zhong
- School of Life Sciences, Henan University, Kaifeng 475000, China
| | - Zhou Guo
- School of Life Sciences, Henan University, Kaifeng 475000, China
| | - Jiahao Yu
- Shandong Zhongbaokang Medical Implements Co., Ltd., Zibo 255000, China
| | - Yuanyuan Ma
- School of Pharmacy, Henan University, Kaifeng 475000, China
| | - Haigang Wu
- School of Artificial Intelligence, Henan University, Zhengzhou 450046, China
| |
Collapse
|
10
|
Zhu J, Gillissen B, Dang Tran DL, May S, Ulrich C, Stockfleth E, Eberle J. Inhibition of Cell Proliferation and Cell Viability by Sinecatechins in Cutaneous SCC Cells Is Related to an Imbalance of ROS and Loss of Mitochondrial Membrane Potential. Antioxidants (Basel) 2022; 11:antiox11071416. [PMID: 35883905 PMCID: PMC9312260 DOI: 10.3390/antiox11071416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/17/2022] [Accepted: 07/18/2022] [Indexed: 02/05/2023] Open
Abstract
The term sinecatechins designates an extract containing a high percentage of catechins obtained from green tea, which is commercially registered as Veregen or Polyphenon E (PE) and may be considered for treatment of cutaneous squamous cell carcinoma (cSCC) and actinic keratosis (AK). As shown here, treatment of four cSCC cell lines with 200 µg/mL of PE resulted in strong, dose-dependent decrease in cell proliferation (20–30%) as well as strongly decreased cell viability (4–21% of controls, 48 h). Effects correlated with loss of mitochondrial membrane potential, whereas early apoptosis was less pronounced. At the protein level, some activation of caspase-3 and enhanced expression of the CDK inhibitor p21 were found. Loss of MMP and induced cell death were, however, largely independent of caspases and of the proapoptotic Bcl-2 proteins Bax and Bak, suggesting that sinecatechins induce also non-apoptotic, alternative cell death pathways, in addition to apoptosis. Reactive oxygen species (ROS) were downregulated in response to PE at 4 h, followed by an increase at 24 h. The contributory role of initially reduced ROS was supported by the antioxidant N-acetyl cysteine, which in combination with PE further enhanced the negative effects on cell viability. Thus, sinecatechins inhibited cell proliferation and viability of cSCC cells, which could suggest the use of PE for AK treatment. The mechanisms appear as linked to an imbalance of ROS levels.
Collapse
Affiliation(s)
- Jiaqi Zhu
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (J.Z.); (D.L.D.T.); (S.M.); (C.U.)
- Department of Gynecology and Obstetrics, Jilin University, Changchun 130001, China
| | - Bernd Gillissen
- Department of Hematology, Oncology, and Tumor Immunology, Charité–Universitätsmedizin Berlin, 13125 Berlin, Germany;
| | - Dieu Linh Dang Tran
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (J.Z.); (D.L.D.T.); (S.M.); (C.U.)
- Beuth-Hochschule für Technik Berlin–University of Applied Sciences, Luxemburger Str. 10, 13353 Berlin, Germany
| | - Stefanie May
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (J.Z.); (D.L.D.T.); (S.M.); (C.U.)
| | - Claas Ulrich
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (J.Z.); (D.L.D.T.); (S.M.); (C.U.)
| | - Eggert Stockfleth
- Dermatologie, Venerologie und Allergologie, Klinikum Bochum, Ruhr-Universität Bochum, Gudrunstr. 56, 44791 Bochum, Germany;
| | - Jürgen Eberle
- Skin Cancer Centre Charité, Department of Dermatology and Allergy, Charité–Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany; (J.Z.); (D.L.D.T.); (S.M.); (C.U.)
- Correspondence: ; Tel.: +49-30-450-518-383
| |
Collapse
|
11
|
Peng X, Ding C, Zhao Y, Hao M, Liu W, Yang M, Xiao F, Zheng Y. Poloxamer 407 and Hyaluronic Acid Thermosensitive Hydrogel-Encapsulated Ginsenoside Rg3 to Promote Skin Wound Healing. Front Bioeng Biotechnol 2022; 10:831007. [PMID: 35866029 PMCID: PMC9294355 DOI: 10.3389/fbioe.2022.831007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Ginsenoside Rg3 has shown beneficial effects in various skin diseases. The current interest in designing and developing hydrogels for biomedical applications continues to grow, inspiring the further development of drug-loaded hydrogels for tissue repair and localized drug delivery. The aim of the present study was to develop an effective and safe hydrogel (Rg3-Gel), using ginsenoside Rg3, and we evaluated the wound-healing potential and therapeutic mechanism of Rg3-Gel. The results indicated that the optimized Rg3-Gel underwent discontinuous phase transition at low and high temperatures. Rg3-Gel also exhibited good network structures, swelling water retention capacity, sustainable release performance, and excellent biocompatibility. Subsequently, the good antibacterial and antioxidant properties of Rg3-Gel were confirmed by in vitro tests. In full-thickness skin defect wounded models, Rg3-Gel significantly accelerated the wound contraction, promoted epithelial and tissue regeneration, and promoted collagen deposition and angiogenesis. In addition, Rg3-Gel increased the expression of autophagy proteins by inhibiting the MAPK and NF-KB pathways in vivo. It simultaneously regulated host immunity by increasing the abundance of beneficial bacteria and the diversity of the wound surface flora. From these preliminary evaluations, it is possible to conclude that Rg3-Gel has excellent application potential in wound-healing drug delivery systems.
Collapse
Affiliation(s)
- Xiaojuan Peng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Chuanbo Ding
- Jilin Agricultural Science and Technology University, Jilin, China
| | - Yingchun Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Mingqian Hao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Wencong Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- *Correspondence: Wencong Liu, ; Min Yang,
| | - Min Yang
- Jilin Agricultural Science and Technology University, Jilin, China
- *Correspondence: Wencong Liu, ; Min Yang,
| | - Fengyan Xiao
- Jilin Agricultural Science and Technology University, Jilin, China
| | - Yinan Zheng
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| |
Collapse
|
12
|
Crucial Role of Reactive Oxygen Species (ROS) for the Proapoptotic Effects of Indirubin Derivatives in Cutaneous SCC Cells. Antioxidants (Basel) 2021; 10:antiox10101514. [PMID: 34679649 PMCID: PMC8532942 DOI: 10.3390/antiox10101514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/31/2021] [Accepted: 09/13/2021] [Indexed: 01/04/2023] Open
Abstract
Efficient drugs are needed for countering the worldwide high incidence of cutaneous squamous cell carcinoma (cSCC) and actinic keratosis. Indirubin derivatives represent promising candidates, but their effects in cSCC cells have not been reported before. Here, we investigated the efficacy of three indirubin derivatives (DKP-071, -073 and -184) in four cSCC cell lines. High efficacy was seen in SCL-I, SCL-II, SCC-12 and SCC-13, resulting in up to 80% loss of cell proliferation, 60% loss of cell viability and 30% induced apoptosis (10 µM). Apoptosis was further enhanced in combinations with TNF-related apoptosis-inducing ligand (TRAIL). Induction of reactive oxygen species (ROS) appeared as critical for these effects. Thus, antioxidative pretreatment completely abolished apoptosis as well as restored cell proliferation and viability. Concerning the pathways, complete activation of caspases cascades (caspases-3, -4, -6, -7, -8 and -9), loss of mitochondrial membrane potential, activation of proapoptotic PKCδ (protein kinase C delta), inhibition of STAT3 (signal transducer and activator of transcription 3), downregulation of antiapoptotic XIAP (X-linked inhibitor of apoptosis protein) and survivin as well as upregulation of the proapoptotic Bcl-2 protein Puma and the cell cycle inhibitor p21 were obtained. Importantly, all activation steps were prevented by antioxidants, thus proving ROS as a master regulator of indirubins' antitumor effects. ROS induction presently develops as an important issue in anticancer therapy.
Collapse
|
13
|
Yu L, Liu J, Zhang TD, Zheng XF, Luo DL, Zhu WL, Qiu XW, Guo LL. Decreased TMEM40 expression is associated with malignant behavior of cutaneous squamous cell carcinoma and inhibits tumor progression. Oncol Lett 2021; 22:606. [PMID: 34188708 PMCID: PMC8227547 DOI: 10.3892/ol.2021.12867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 03/08/2021] [Indexed: 01/22/2023] Open
Abstract
Cutaneous squamous cell carcinoma (CSCC) is one of the most common types of skin cancer in humans worldwide. The identification and characterization of cancer-associated transmembrane proteins are important for understanding the molecular biology of CSCC. The aim of the present study was to evaluate the expression pattern of transmembrane protein 40 (TMEM40) in CSCC and its clinical significance. The underlying mechanisms were also examined. Reverse transcription-quantitative PCR, western blot and immunohistochemistry analysis were used to determine the relative expression of TMEM40 in CSCC cell lines and clinical tissue samples. The effect of TMEM40 gene silencing on cell proliferation was also evaluated using Cell Counting Kit-8 assays. Wound healing assays, flow cytometry and Transwell assays were used to explore the migration, cell cycle distribution/apoptosis and invasion of CSCC cells following TMEM40 silencing, respectively. In the present study, increased TMEM40 expression was observed in CSCC tissue samples, compared with normal skin, and TMEM40 expression was associated with large tumor size in patients with CSCC. In vitro functional assays indicated that TMEM40 was involved in the regulation of A431 and SCL1 cell growth through its effects on the cell cycle and apoptosis. Silencing TMEM40 in A431 and SCL1 cells resulted in cell cycle arrest at the G0/G1 phase and promoted apoptosis. In addition, migration and invasion were significantly inhibited following silencing of TMEM40 expression in CSCC cells. Taken together, the results of the present study indicated that reduced TMEM40 expression could inhibit CSCC development and that TMEM40 may represent a therapeutic target in CSCC.
Collapse
Affiliation(s)
- Lei Yu
- Department of Dermatology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Jie Liu
- Department of Clinical Laboratory, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Tang-De Zhang
- Department of Dermatology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Xiu-Fen Zheng
- Department of Dermatology, Shunde Hospital of Southern Medical University, Shunde, Guangdong 528308, P.R. China
| | - Dong-Lan Luo
- Department of Dermatology, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong 518028, P.R. China
| | - Wei-Liang Zhu
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Xian-Wen Qiu
- Department of Dermatology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Lin-Lang Guo
- Department of Pathology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
14
|
Tartaglione MF, Eléxpuru Zabaleta M, Lazzarini R, Piva F, Busilacchi EM, Poloni A, Ledda C, Rapisarda V, Santarelli L, Bracci M. Apoptotic mechanism activated by blue light and cisplatinum in cutaneous squamous cell carcinoma cells. Int J Mol Med 2021; 47:48. [PMID: 33576463 PMCID: PMC7891828 DOI: 10.3892/ijmm.2021.4881] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 01/18/2021] [Indexed: 01/05/2023] Open
Abstract
New approaches are being studied for the treatment of skin cancer. It has been reported that light combined with cisplatinum may be effective against skin cancer. In the present study, the effects of specific light radiations and cisplatinum on A431 cutaneous squamous cell carcinoma (cSCC) and HaCaT non-tumorigenic cell lines were investigated. Both cell lines were exposed to blue and red light sources for 3 days prior to cisplatinum treatment. Viability, apoptosis, cell cycle progression and apoptotic-related protein expression levels were investigated. The present results highlighted that combined treatment with blue light and cisplatinum was more effective in reducing cell viability compared with single treatments. Specifically, an increase in the apoptotic rate was observed when the cells were treated with blue light and cisplatinum, as compared to treatment with blue light or cisplatinum alone. Combined treatment with blue light and cisplatinum also caused cell cycle arrest at the S phase. Treatment with cisplatinum following light exposure induced the expression of apoptotic proteins in the A431 and HaCaT cell lines, which tended to follow different apoptotic mechanisms. On the whole, these data indicate that blue light combined with cisplatinum may be a promising treatment for cSCC.
Collapse
Affiliation(s)
- Maria Fiorella Tartaglione
- Section of Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, I-60126 Ancona, Italy
| | - María Eléxpuru Zabaleta
- Section of Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, I-60126 Ancona, Italy
| | - Raffaella Lazzarini
- Section of Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, I-60126 Ancona, Italy
| | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, I-60131 Ancona, Italy
| | - Elena Marinelli Busilacchi
- Section of Hematology, Department of Clinical and Molecular Science, Polytechnic University of Marche, I-60126 Ancona, Italy
| | - Antonella Poloni
- Section of Hematology, Department of Clinical and Molecular Science, Polytechnic University of Marche, I-60126 Ancona, Italy
| | - Caterina Ledda
- Section of Occupational Medicine, Department of Clinical and Experimental Medicine, University of Catania, I-95124 Catania, Italy
| | - Venerando Rapisarda
- Section of Occupational Medicine, Department of Clinical and Experimental Medicine, University of Catania, I-95124 Catania, Italy
| | - Lory Santarelli
- Section of Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, I-60126 Ancona, Italy
| | - Massimo Bracci
- Section of Occupational Medicine, Department of Clinical and Molecular Sciences, Polytechnic University of Marche, I-60126 Ancona, Italy
| |
Collapse
|
15
|
Antifibrotic effects of Hypocrellin A combined with LED red light irradiation on keloid fibroblasts by counteracting the TGF-β/Smad/autophagy/apoptosis signalling pathway. Photodiagnosis Photodyn Ther 2021; 34:102202. [PMID: 33556618 DOI: 10.1016/j.pdpdt.2021.102202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/21/2021] [Accepted: 01/25/2021] [Indexed: 11/23/2022]
Abstract
Keloids are characterized by abnormal proliferation of fibroblasts and continuous deposition of extracellular matrix (ECM) components. In the field of dermopathy, photodynamic therapy (PDT) with visible light has been increasingly investigated. The natural photosensitizer Hypocrellin A (HA) was shown to have excellent light induced anticancer, antimicrobial and antiviral activities. In this experiment, we investigated the impacts of HA united light-emitting diode (LED) red light irradiation on human keloid fibroblast cells (KFs). Our results showed that HA combined with red light irradiation treatment (HA-R-PDT) decreased KF viability, reduced KF collagen production and ECM accumulation, inhibited cell proliferation, suppressed cell invasion and induced cell apoptosis. Moreover, our observations demonstrated that the TGF-β/Smad signalling pathway and autophagy were restrained by HA-R-PDT. TGF-β1 could promote autophagy in KFs through both the Smad and ERK pathways, while inhibition of autophagy altered the TGF-β1 levels through negative feedback. Therefore, HA-R-PDT suppressed cell hyperproliferation, collagen synthesis and ECM accumulation of KFs by regulating the TGF-β1-ERK-autophagy-apoptosis signalling pathway. HA-R-PDT deserves systematic investigation as a potential therapeutic strategy for keloids, and autophagy might be a promising candidate in the treatment of KFs.
Collapse
|
16
|
Dong H, Yao X, Guo L, Yu S, Duan W, Liu W, Wang X. Purification of hypocrellins from
Shiraia bambusicola
by coordinated high‐speed countercurrent chromatography using cupric chloride as a complexing agent. J Sep Sci 2021; 44:1383-1390. [DOI: 10.1002/jssc.202001133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/29/2020] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Hongjing Dong
- School of Pharmaceutical Sciences Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province Shandong Analysis and Test Center Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
- Shandong Provincial Collaborative innovation Center for Quality Control and Construction of the Whole Industrial Chain of Traditional Chinese Medicine Jinan P. R. China
| | - Xue Yao
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province Shandong Analysis and Test Center Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
| | - Lanping Guo
- Resource Center of Chinese Materia Medica State Key Laboratory Breeding Base of Dao‐di Herbs China Academy of Chinese Medical Sciences Beijing P. R. China
| | - Shuai Yu
- School of Pharmaceutical Sciences Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province Shandong Analysis and Test Center Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
| | - Wenjuan Duan
- School of Pharmaceutical Sciences Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province Shandong Analysis and Test Center Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
| | - Wei Liu
- School of Pharmaceutical Sciences Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province Shandong Analysis and Test Center Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
| | - Xiao Wang
- School of Pharmaceutical Sciences Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
- Key Laboratory for Applied Technology of Sophisticated Analytical Instruments of Shandong Province Shandong Analysis and Test Center Qilu University of Technology (Shandong Academy of Sciences) Jinan P. R. China
| |
Collapse
|
17
|
Xin Y, Zou L, Lang S. 4-Octyl itaconate (4-OI) attenuates lipopolysaccharide-induced acute lung injury by suppressing PI3K/Akt/NF-κB signaling pathways in mice. Exp Ther Med 2021; 21:141. [PMID: 33456508 PMCID: PMC7791918 DOI: 10.3892/etm.2020.9573] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022] Open
Abstract
The progression of acute lung injury (ALI) is attributable to inflammation and oxidative stress. The cell-permeable itaconate analog 4-octyl itaconate (4-OI) provides protection against inflammatory responses and oxidative stress. However, whether 4-OI can protect against ALI remains poorly understood. The aim of this study was to explore the protective effects of 4-OI against LPS-induced ALI and the underlying mechanisms using hematoxylin and eosin (H&E) to observe lung morphology, ELISA and reverse transcription-quantitative PCR to measure the levels of IL-1β, TNF-α and IL-6 and western blotting to examine the levels of PI3K, Akt and NF-κB. The present study demonstrates that intraperitoneal administration of 4-OI (25 mg/kg) 2 h before lipopolysaccharide (LPS; 5 mg/kg) intratracheal injection significantly alleviated the lung tissue injury induced by LPS, reducing the production of proinflammatory cytokines and reactive oxygen species (ROS) in vivo. Furthermore, 4-OI and the antioxidant N-acetyl-L-cysteine markedly suppressed PI3K and Akt phosphorylation in LPS-treated RAW264.7 macrophage cells in vitro. Further study demonstrated that a pharmacological inhibitor of the phosphoinositide 3-kinase (PI3K)-Akt pathway, LY294002, inhibited the expression of NF-κB p65 in the nuclear fraction and decreased the production of inflammatory cytokines. Collectively, the experimental results of the present study provide evidence that 4-OI significantly decreased LPS-induced lung inflammation by suppressing ROS-mediated PI3K/Akt/NF-κB signaling pathways. These results suggest that 4-OI could be a valuable therapeutic drug in the treatment of ALI.
Collapse
Affiliation(s)
- Yan Xin
- Department of Anesthesiology, Changchun Maternity Hospital, Changchun, Jilin 130042, P.R. China
| | - Lili Zou
- Department of Anesthesiology, General Hospital of Ning Xia Medical University, Yin Chuan, Ningxia 750004, P.R. China
| | - Shuhui Lang
- Department of Anesthesiology, General Hospital of Ning Xia Medical University, Yin Chuan, Ningxia 750004, P.R. China
| |
Collapse
|
18
|
Martins WK, Belotto R, Silva MN, Grasso D, Suriani MD, Lavor TS, Itri R, Baptista MS, Tsubone TM. Autophagy Regulation and Photodynamic Therapy: Insights to Improve Outcomes of Cancer Treatment. Front Oncol 2021; 10:610472. [PMID: 33552982 PMCID: PMC7855851 DOI: 10.3389/fonc.2020.610472] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 12/03/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer is considered an age-related disease that, over the next 10 years, will become the most prevalent health problem worldwide. Although cancer therapy has remarkably improved in the last few decades, novel treatment concepts are needed to defeat this disease. Photodynamic Therapy (PDT) signalize a pathway to treat and manage several types of cancer. Over the past three decades, new light sources and photosensitizers (PS) have been developed to be applied in PDT. Nevertheless, there is a lack of knowledge to explain the main biochemical routes needed to trigger regulated cell death mechanisms, affecting, considerably, the scope of the PDT. Although autophagy modulation is being raised as an interesting strategy to be used in cancer therapy, the main aspects referring to the autophagy role over cell succumbing PDT-photoinduced damage remain elusive. Several reports emphasize cytoprotective autophagy, as an ultimate attempt of cells to cope with the photo-induced stress and to survive. Moreover, other underlying molecular mechanisms that evoke PDT-resistance of tumor cells were considered. We reviewed the paradigm about the PDT-regulated cell death mechanisms that involve autophagic impairment or boosted activation. To comprise the autophagy-targeted PDT-protocols to treat cancer, it was underlined those that alleviate or intensify PDT-resistance of tumor cells. Thereby, this review provides insights into the mechanisms by which PDT can be used to modulate autophagy and emphasizes how this field represents a promising therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Waleska K Martins
- Laboratory of Cell and Membrane, Anhanguera University of São Paulo, São Paulo, Brazil
| | - Renata Belotto
- Perola Byington Hospital Gynecology - Lasertherapy Clinical Research Department, São Paulo, Brazil
| | - Maryana N Silva
- Laboratory of Cell and Membrane, Anhanguera University of São Paulo, São Paulo, Brazil
| | - Daniel Grasso
- CONICET, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maynne D Suriani
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Brazil
| | - Tayná S Lavor
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Brazil
| | - Rosangela Itri
- Institute of Physics, University of São Paulo, São Paulo, Brazil
| | | | - Tayana M Tsubone
- Institute of Chemistry, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
19
|
Babaei G, Aziz SGG, Jaghi NZZ. EMT, cancer stem cells and autophagy; The three main axes of metastasis. Biomed Pharmacother 2020; 133:110909. [PMID: 33227701 DOI: 10.1016/j.biopha.2020.110909] [Citation(s) in RCA: 268] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/29/2020] [Accepted: 10/17/2020] [Indexed: 02/07/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) and Cancer stem-like cells (CSCs) are major factors contributing to the metastasis of cancer cells. Consequently, the signaling pathways involved in both processes are appropriate therapeutic targets in the treatment of metastasis. Autophagy is another process that has recently attracted the attention of many researchers; depending on the type of cancer and tissue and the stage of cancer, this process can play a dual role in the development of cancer cells. Studies on cancer cells have shown that different signaling pathways are involved in all three processes, namely, cancer stem cells, autophagy, and EMT. The purpose of this study was to investigate and elucidate the relationship between the effective signaling pathways in all three processes, which could play an effective role in determining appropriate therapeutic goals.
Collapse
Affiliation(s)
- Ghader Babaei
- Department of Biochemistry, Faculty of Medicine, Urmia University Medical Sciences (UMSU), Urmia, Iran; Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran.
| | | | - Nasrin Zare Zavieyh Jaghi
- Department of Biochemistry, Faculty of Medicine, Urmia University Medical Sciences (UMSU), Urmia, Iran
| |
Collapse
|
20
|
Keerthiga R, Zhao Z, Pei D, Fu A. Photodynamic Nanophotosensitizers: Promising Materials for Tumor Theranostics. ACS Biomater Sci Eng 2020; 6:5474-5485. [PMID: 33320544 DOI: 10.1021/acsbiomaterials.0c01058] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Photodynamic theranostics/therapy (PDT) is a potential strategy for selectively imaging malignant sites and treating cancer via a non-invasive therapeutic method. Photosensitizers, the crucial components of PDT, enable colocalization of photons and light, and photon/light therapy in the therapeutic window of 400-900 nm exhibits photocytotoxicity to tumor cells. Due to their high biostability and photocytotoxicity, nanophotosensitizers (NPSs) are of much interest for malignant tumor theranostics at present. NPS-activated photons transfer energy through the absorption of a photon and convert molecular oxygen to the singlet reactive oxygen species, which leads to apoptosis and necrosis. Moreover, NPSs modified by polymers, including PLGA, PEG-PLA, PDLLA, PVCL-g-PLA, and P(VCL-co-VIM)-g-PLA, exhibit excellent biocompatibility, and a tumor-targeting molecule linked on the nanoparticle surface can precisely deliver NPSs into the tumor region. The development of NPSs will accelerate the progress in tumor theranostics through the photon/light pathway.
Collapse
Affiliation(s)
- Rajendiran Keerthiga
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zizhen Zhao
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Desheng Pei
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing 400714, China
| | - Ailing Fu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|