Redox in Cancer Occurence and Therapy

A special issue of Antioxidants (ISSN 2076-3921). This special issue belongs to the section "Health Outcomes of Antioxidants and Oxidative Stress".

Deadline for manuscript submissions: closed (15 February 2022) | Viewed by 23381

Special Issue Editors


E-Mail Website
Guest Editor
Institute of Health and Medical Technology, Hefei Institutes of Physical Sciences, Chinese Academy of Sciences, Hefei, China
Interests: non-thermal plasma; free radicals; cell death; anti-tumor immunity; radiotherapy
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Department of Physics, City University of Hong Kong, Tat Chee Avenue, Kowloon Tong, Kowloon, Hong Kong, China
Interests: radiation biology; radiation physics; radiation protection; radiation dosimetry; computer simulation

Special Issue Information

Dear colleagues,

As we know, oxidation and reduction of molecules are commonly associated with various processes of physiology and pathology. In particular, the oxidative stress and the corresponding activation of anti-oxidative systems have been regarded as essential and necessary in gene mutation, malignant transformation, tumor formation and development, cell death, and inflammation-type response in cancer treatment. Therefore, understanding the important role of redox in the occurrence and treatment of cancer from different aspects will provide considerable insights into the prevention of cancer, optimization of treatments, improvement of prognosis, and relief of side effects.  

In this Special Issue, we look forward to collecting research papers which explore the role of redox in different research fields of tumorigenesis and oncotherapy, including chemical, physical, and biological aspects, and in particular non-conventional aspects. We also welcome contributions of systemic reviews which survey and summarize new advances in these fields.

Prof. Dr. Wei Han
Prof. Dr. Peter K.N. Yu
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Antioxidants is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • redox
  • tumorigenesis
  • oncotherapy
  • cell death
  • inflammation

Published Papers (6 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

20 pages, 3464 KiB  
Article
Single-Cell Transcriptome Analysis of Radiation Pneumonitis Mice
by Miaomiao Yang, Qiang Fan, Tom K. Hei, Guodong Chen, Wei Cao, Gang Meng and Wei Han
Antioxidants 2022, 11(8), 1457; https://doi.org/10.3390/antiox11081457 - 26 Jul 2022
Cited by 7 | Viewed by 4073
Abstract
Radiation-induced lung injury (RILI), especially radiation pneumonitis (RP), is a common clinical complication associated with thoracic radiotherapy for malignant tumors. However, the specific contributions of each cell subtype to this process are unknown. Here, we provide the single-cell pathology landscape of the RP [...] Read more.
Radiation-induced lung injury (RILI), especially radiation pneumonitis (RP), is a common clinical complication associated with thoracic radiotherapy for malignant tumors. However, the specific contributions of each cell subtype to this process are unknown. Here, we provide the single-cell pathology landscape of the RP in a mouse model by unbiased single-cell RNA-seq (scRNA-seq). We found a decline of type 2 alveolar cells in the RP lung tissue, with an expansion of macrophages, especially the Fabp4low and Spp1high subgroup, while Fabp4high macrophages were almost depleted. We observed an elevated expression of multiple mitochondrial genes in the RP group, indicating a type 2 alveolar cell (AT2) response to oxidative stress. We also calculated the enrichment of a cGAS-STING signaling pathway, which may be involved in regulating inflammatory responses and cancer progression in AT2 cells of PR mice. We delineate markers and transcriptional states, identify a type 2 alveolar cell, and uncover fundamental determinants of lung fibrosis and inflammatory response in RP lung tissue of mice. Full article
(This article belongs to the Special Issue Redox in Cancer Occurence and Therapy)
Show Figures

Figure 1

17 pages, 1821 KiB  
Article
Combined Activity of the Redox-Modulating Compound Setanaxib (GKT137831) with Cytotoxic Agents in the Killing of Acute Myeloid Leukemia Cells
by Muhammed Burak Demircan, Peter C. Mgbecheta, Anne Kresinsky, Tina M. Schnoeder, Katrin Schröder, Florian H. Heidel and Frank D. Böhmer
Antioxidants 2022, 11(3), 513; https://doi.org/10.3390/antiox11030513 - 8 Mar 2022
Cited by 4 | Viewed by 3240
Abstract
Acute myeloid leukemia (AML) cells harbor elevated levels of reactive oxygen species (ROS), which promote cell proliferation and cause oxidative stress. Therefore, the inhibition of ROS formation or elevation beyond a toxic level have been considered as therapeutic strategies. ROS elevation has recently [...] Read more.
Acute myeloid leukemia (AML) cells harbor elevated levels of reactive oxygen species (ROS), which promote cell proliferation and cause oxidative stress. Therefore, the inhibition of ROS formation or elevation beyond a toxic level have been considered as therapeutic strategies. ROS elevation has recently been linked to enhanced NADPH oxidase 4 (NOX4) activity. Therefore, the compound Setanaxib (GKT137831), a clinically advanced ROS-modulating substance, which has initially been identified as a NOX1/4 inhibitor, was tested for its inhibitory activity on AML cells. Setanaxib showed antiproliferative activity as single compound, and strongly enhanced the cytotoxic action of anthracyclines such as daunorubicin in vitro. Setanaxib attenuated disease in a mouse model of FLT3-ITD driven myeloproliferation in vivo. Setanaxib did not significantly inhibit FLT3-ITD signaling, including FLT3 autophosphorylation, activation of STAT5, AKT, or extracellular signal regulated kinase 1 and 2 (ERK1/2). Surprisingly, the effects of Setanaxib on cell proliferation appeared to be independent of the presence of NOX4 and were not associated with ROS quenching. Instead, Setanaxib caused elevation of ROS levels in the AML cells and importantly, enhanced anthracycline-induced ROS formation, which may contribute to the combined effects. Further assessment of Setanaxib as potential enhancer of cytotoxic AML therapy appears warranted. Full article
(This article belongs to the Special Issue Redox in Cancer Occurence and Therapy)
Show Figures

Graphical abstract

12 pages, 2338 KiB  
Article
Sulforaphane-Mediated Nrf2 Activation Prevents Radiation-Induced Skin Injury through Inhibiting the Oxidative-Stress-Activated DNA Damage and NLRP3 Inflammasome
by Jinlong Wei, Qin Zhao, Yuyu Zhang, Weiyan Shi, Huanhuan Wang, Zhuangzhuang Zheng, Lingbin Meng, Ying Xin and Xin Jiang
Antioxidants 2021, 10(11), 1850; https://doi.org/10.3390/antiox10111850 - 22 Nov 2021
Cited by 32 | Viewed by 3398
Abstract
This article mainly observed the protective effect of sulforaphane (SFN) on radiation-induced skin injury (RISI). In addition, we will discuss the mechanism of SFN’s protection on RISI. The RISI model was established by the irradiation of the left thigh under intravenous anesthesia. Thirty-two [...] Read more.
This article mainly observed the protective effect of sulforaphane (SFN) on radiation-induced skin injury (RISI). In addition, we will discuss the mechanism of SFN’s protection on RISI. The RISI model was established by the irradiation of the left thigh under intravenous anesthesia. Thirty-two C57/BL6 mice were randomly divided into control group (CON), SFN group, irradiation (IR) group, and IR plus SFN (IR/SFN) group. At eight weeks after irradiation, the morphological changes of mouse skin tissues were detected by H&E staining. Then, the oxidative stress and inflammatory response indexes in mouse skin tissues, as well as the expression of Nrf2 and its downstream antioxidant genes, were evaluated by ELISA, real-time PCR, and Western blotting. The H&E staining showed the hyperplasia of fibrous tissue in the mouse dermis and hypodermis of the IR group. Western blotting and ELISA results showed that the inflammasome of NLRP3, caspase-1, and IL-1β, as well as oxidative stress damage indicators ROS, 4-HNE, and 3-NT, in the skin tissues of mice in the IR group were significantly higher than those in the control group (p < 0.05). However, the above pathological changes declined sharply after SFN treatment (p < 0.05). In addition, the expressions of Nrf2 and its regulated antioxidant enzymes, including CAT and HO-1, were higher in the skin tissues of SFN and IR/SFN groups, but lower in the control and IR groups (p < 0.05). SFN may be able to suppress the oxidative stress by upregulating the expression and function of Nrf2, and subsequently inhibiting the activation of NLRP3 inflammasome and DNA damage, so as to prevent and alleviate the RISI. Full article
(This article belongs to the Special Issue Redox in Cancer Occurence and Therapy)
Show Figures

Figure 1

Review

Jump to: Research

17 pages, 1553 KiB  
Review
Prospective Application of Ferroptosis in Hypoxic Cells for Tumor Radiotherapy
by Jing Su, Qin Zhao, Zhuangzhuang Zheng, Huanhuan Wang, Chenbin Bian, Lingbin Meng, Ying Xin and Xin Jiang
Antioxidants 2022, 11(5), 921; https://doi.org/10.3390/antiox11050921 - 7 May 2022
Cited by 20 | Viewed by 3944
Abstract
Radiation therapy plays an increasingly important role in cancer treatment. It can inhibit the progression of various cancers through radiation-induced DNA breakage and reactive oxygen species (ROS) overload. Unfortunately, solid tumors, such as breast and lung cancer, often develop a hypoxic microenvironment due [...] Read more.
Radiation therapy plays an increasingly important role in cancer treatment. It can inhibit the progression of various cancers through radiation-induced DNA breakage and reactive oxygen species (ROS) overload. Unfortunately, solid tumors, such as breast and lung cancer, often develop a hypoxic microenvironment due to insufficient blood supply and rapid tumor proliferation, thereby affecting the effectiveness of radiation therapy. Restraining hypoxia and improving the curative effect of radiotherapy have become difficult problems. Ferroptosis is a new type of cell death caused by lipid peroxidation due to iron metabolism disorders and ROS accumulation. It plays an important role in both hypoxia and radiotherapy and can enhance the radiosensitivity of hypoxic tumor cells by amplifying oxidative stress or inhibiting antioxidant regulation. In this review, we summarize the internal relationship and related mechanisms between ferroptosis and hypoxia, thus exploring the possibility of inducing ferroptosis to improve the prognosis of hypoxic tumors. Full article
(This article belongs to the Special Issue Redox in Cancer Occurence and Therapy)
Show Figures

Figure 1

22 pages, 2164 KiB  
Review
Oxidative Stress in Cancer Immunotherapy: Molecular Mechanisms and Potential Applications
by Ruolan Liu, Liyuan Peng, Li Zhou, Zhao Huang, Chengwei Zhou and Canhua Huang
Antioxidants 2022, 11(5), 853; https://doi.org/10.3390/antiox11050853 - 27 Apr 2022
Cited by 10 | Viewed by 3681
Abstract
Immunotherapy is an effective treatment option that revolutionizes the management of various cancers. Nevertheless, only a subset of patients receiving immunotherapy exhibit durable responses. Recently, numerous studies have shown that oxidative stress induced by reactive oxygen species (ROS) plays essential regulatory roles in [...] Read more.
Immunotherapy is an effective treatment option that revolutionizes the management of various cancers. Nevertheless, only a subset of patients receiving immunotherapy exhibit durable responses. Recently, numerous studies have shown that oxidative stress induced by reactive oxygen species (ROS) plays essential regulatory roles in the tumor immune response, thus regulating immunotherapeutic effects. Specifically, studies have revealed key roles of ROS in promoting the release of tumor-associated antigens, manipulating antigen presentation and recognition, regulating immune cell phenotypic differentiation, increasing immune cell tumor infiltration, preventing immune escape and diminishing immune suppression. In the present study, we briefly summarize the main classes of cancer immunotherapeutic strategies and discuss the interplay between oxidative stress and anticancer immunity, with an emphasis on the molecular mechanisms underlying the oxidative stress-regulated treatment response to cancer immunotherapy. Moreover, we highlight the therapeutic opportunities of manipulating oxidative stress to improve the antitumor immune response, which may improve the clinical outcome. Full article
(This article belongs to the Special Issue Redox in Cancer Occurence and Therapy)
Show Figures

Figure 1

25 pages, 1160 KiB  
Review
Mechanism, Prevention, and Treatment of Radiation-Induced Salivary Gland Injury Related to Oxidative Stress
by Zijing Liu, Lihua Dong, Zhuangzhuang Zheng, Shiyu Liu, Shouliang Gong, Lingbin Meng, Ying Xin and Xin Jiang
Antioxidants 2021, 10(11), 1666; https://doi.org/10.3390/antiox10111666 - 22 Oct 2021
Cited by 14 | Viewed by 3819
Abstract
Radiation therapy is a common treatment for head and neck cancers. However, because of the presence of nerve structures (brain stem, spinal cord, and brachial plexus), salivary glands (SGs), mucous membranes, and swallowing muscles in the head and neck regions, radiotherapy inevitably causes [...] Read more.
Radiation therapy is a common treatment for head and neck cancers. However, because of the presence of nerve structures (brain stem, spinal cord, and brachial plexus), salivary glands (SGs), mucous membranes, and swallowing muscles in the head and neck regions, radiotherapy inevitably causes damage to these normal tissues. Among them, SG injury is a serious adverse event, and its clinical manifestations include changes in taste, difficulty chewing and swallowing, oral infections, and dental caries. These clinical symptoms seriously reduce a patient’s quality of life. Therefore, it is important to clarify the mechanism of SG injury caused by radiotherapy. Although the mechanism of radiation-induced SG injury has not yet been determined, recent studies have shown that the mechanisms of calcium signaling, microvascular injury, cellular senescence, and apoptosis are closely related to oxidative stress. In this article, we review the mechanism by which radiotherapy causes oxidative stress and damages the SGs. In addition, we discuss effective methods to prevent and treat radiation-induced SG damage. Full article
(This article belongs to the Special Issue Redox in Cancer Occurence and Therapy)
Show Figures

Graphical abstract

Back to TopTop