1
|
Craige SM, Kaur G, Bond JM, Caliz AD, Kant S, Keaney JF. Endothelial Reactive Oxygen Species: Key Players in Cardiovascular Health and Disease. Antioxid Redox Signal 2025; 42:905-932. [PMID: 39213161 PMCID: PMC12183504 DOI: 10.1089/ars.2024.0706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Significance: Endothelial cells (ECs) line the entire vasculature system and serve as both barriers and facilitators of intra- and interorgan communication. Positioned to rapidly sense internal and external stressors, ECs dynamically adjust their functionality. Endothelial dysfunction occurs when the ability of ECs to react to stressors is impaired, which precedes many cardiovascular diseases (CVDs). While EC reactive oxygen species (ROS) have historically been implicated as mediators of endothelial dysfunction, more recent studies highlight the central role of ROS in physiological endothelial signaling. Recent Advances: New evidence has uncovered that EC ROS are fundamental in determining how ECs interact with their environment and respond to stress. EC ROS levels are mediated by external factors such as diet and pathogens, as well as inherent characteristics, including sex and location. Changes in EC ROS impact EC function, leading to changes in metabolism, cell communication, and potentially disrupted signaling in CVDs. Critical Issues: Current endothelial biology concepts integrate the dual nature of ROS, emphasizing the importance of EC ROS in physiological stress adaptation and their contribution to CVDs. Understanding the discrete, localized signaling of EC ROS will be critical in preventing adverse cardiovascular outcomes. Future Directions: Exploring how the EC ROS environment alters EC function and cross-cellular communication is critical. Considering the inherent heterogeneity among EC populations and understanding how EC ROS contribute to this diversity and the role of sexual dimorphism in the EC ROS environment will be fundamental for developing new effective cardiovascular treatment strategies. Antioxid. Redox Signal. 42, 905-932.
Collapse
Affiliation(s)
- Siobhan M. Craige
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia, USA
| | - Gaganpreet Kaur
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob M. Bond
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia, USA
- Translational Biology, Medicine, and Health Program, Virginia Tech, Roanoke, Virginia, USA
| | - Amada D. Caliz
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shashi Kant
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John F. Keaney
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Shi X, Shen T, Gu M, Guan Y, Aimaiti G, Yu W, Zhang X, Yuan WE, Su J. Development of a novel Cu-Mn hydroxide layered nanosheet-loaded drug modulating the tumour microenvironment and enhancing antitumor effects. J Colloid Interface Sci 2025; 696:137904. [PMID: 40393131 DOI: 10.1016/j.jcis.2025.137904] [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: 02/13/2025] [Revised: 05/13/2025] [Accepted: 05/15/2025] [Indexed: 05/22/2025]
Abstract
The tumor microenvironment (TME) impedes the effectiveness of therapeutic strategies such as chemodynamic therapy (CDT). This study presents a novel nanoscale drug delivery system designed for the precise release of the chemotherapeutic agent doxorubicin (DOX), aiming to overcome treatment limitations, reduce systemic toxicity, and enhance antitumor efficacy. Mn(III) serves as an immunomodulatory agent, while Cu(II) regulates the levels of glutathione (GSH). Layered double hydroxides (LDHs) were synthesized and efficiently loaded with DOX, followed by surface modification with hyaluronic acid (HA). The HA-coated LDH/DOX nanocarriers showed effective internalization by tumor cells and provided a pH-responsive release of DOX. In vitro, the LDH/HA/DOX complex exhibited strong catalytic activity in the Fenton reaction. In vivo studies using an H22 hepatocarcinoma model confirmed its potent antitumor activity and excellent biocompatibility. Immunohistochemical analyses revealed that treatment with LDH/HA/DOX significantly increased infiltration of M1-polarized tumor-associated macrophages (TAMs), CD4 + T cells and CD8 + T cells, while decreasing M2-polarized TAMs. This change in immune cell profile was associated with notable tumor growth inhibition.
Collapse
Affiliation(s)
- Xiaoying Shi
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Tianyi Shen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Muge Gu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Yuanye Guan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Gulizeba Aimaiti
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Wei Yu
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Xiangqi Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China
| | - Wei-En Yuan
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China.
| | - Jing Su
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai 200240, China; Inner Mongolia Research Institute of Shanghai Jiao Tong University, China.
| |
Collapse
|
4
|
Xiong B, Zhang Y, Liu S, Liao S, Zhou Z, He Q, Zhou Y. NOX Family: Regulators of Reactive Oxygen Species Balance in Tumor Cells. FASEB J 2025; 39:e70565. [PMID: 40266050 PMCID: PMC12017260 DOI: 10.1096/fj.202500238rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Cancer cells are capable of surviving, proliferating, and invading or migrating within hypoxic environments by regulating various adaptive mechanisms. Due to the activation of oncogenes and the inactivation of tumor suppressor genes, and relative deficiencies in oxygen and nutrients, cancer cells demonstrate elevated production of reactive oxygen species (ROS), primarily sourced from NADPH oxidases (NOX family). A key aspect of the reorientation of tumor cell metabolism is the combating of cellular oxidative stress through the promotion of antioxidant molecule synthesis to counteract ROS production. Given that most cancers experience hypoxia and that NOX is closely linked to numerous redox-dependent signaling pathways, the expression and function of NOX are altered in various malignancies. Therefore, this review summarizes the characteristics of NOX family members, their influence on tumor proliferation, invasion, and migration, the role of NOX in promoting tumor angiogenesis, the impact of NOX on the function of immune cells within the tumor microenvironment, and the potential of targeting NOX in tumor therapy. This aims to offer a fresh viewpoint on a comprehensive understanding of the functions of NOX family members.
Collapse
Affiliation(s)
- Bin Xiong
- Department of Radiation OncologyThe Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer HospitalChangshaHunanChina
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| | - Yang Zhang
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| | - Siyi Liu
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| | - Shan Liao
- Department of PathologyThe Third Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Zihua Zhou
- Department of OncologyLoudi Central HospitalLoudiHunanChina
| | - Qian He
- Department of Radiation OncologyThe Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer HospitalChangshaHunanChina
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| |
Collapse
|
5
|
Ghosh R, Bhowmik A, Biswas S, Samanta P, Sarkar R, Pakhira S, Mondal M, Hajra S, Saha P. Natural flavonoid Orientin restricts 5-Fluorouracil induced cancer stem cells mediated angiogenesis by regulating HIF1α and VEGFA in colorectal cancer. Mol Med 2025; 31:85. [PMID: 40045186 PMCID: PMC11881437 DOI: 10.1186/s10020-024-01032-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/06/2024] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Cancer stem cells are a small subpopulation of cells which are responsible for tumor metastasis, angiogenesis, drug resistance etc. 5-Fluorouracil (5FU), a common therapeutic drug used in colorectal cancer treatment is reported to enrich CSCs, tumor recurrence and induces severe organ toxicities resulting in poor clinical outcome in patients. Therefore, we introduced a natural flavonoid Orientin in combination with 5FU to mitigate the CSC mediated angiogenesis and induced toxicities. METHODS Tumorosphere generation, flow cytometry, immunofluorescence assay, and western blotting were performed by using 5FU and Orientin individually and both treated colorectal cells and CSCs. In silico study was carried out to check the interaction between HIF1α and Orientin. In ovo chorioallantoic membrane (CAM) assay and tube formation assay using HUVECs were performed to monitor CSC mediated angiogenesis. In vivo CT26 syngeneic mice model was used to validate in silico and ex vivo results. RESULTS We found that 5FU treatment significantly increased the CD44+/CD133+ CSC population. In contrast, this CSC population in CSC enriched spheres (CES) derived from HCT116 cells were decreased by combination of Orientin and 5FU. Decrease of CSC's stemness properties was also noted, as evidenced by the downregulation of NANOG, SOX2 and OCT4. This new therapeutic strategy also inhibited CSC mediated angiogenesis by downregulating 5FU induced ROS, NO and LPO in those tumorospheres. Combination of Orientin and 5FU significantly reduced CSC mediated angiogenesis in HUVEC and CAM. Additionally, in silico study predicted that Orientin can bind to the PAS domain of HIF1α, a crucial factor for promoting angiogenesis. Expression of HIF1α and VEGFA were also decreased when the CESs were exposed to the combinatorial treatment. Additionally, we found that treatment with 5FU alone resulted reduction in tumor volume but it enriched CSCs and produced nephrotoxicity and hepatotoxicity in vivo. Combined treatment also considerably reduced the CD44+/CD133+ CSC population and hindered angiogenesis in a therapeutic in vivo model in BALB/c mice. CONCLUSIONS This novel treatment strategy of "Orientin with 5FU" is likely to improve the efficiency of conventional chemotherapy and may suppress disease recurrence in colorectal cancer by limiting CSC mediated angiogenesis.
Collapse
Affiliation(s)
- Rituparna Ghosh
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Arijit Bhowmik
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India.
| | - Souradeep Biswas
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Priya Samanta
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Rupali Sarkar
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Shampa Pakhira
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Mrinmoyee Mondal
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Subhadip Hajra
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India.
| |
Collapse
|
6
|
Hyun J, Lee SY, An J, Lee YB, Bhang SH. Strengthening the cellular function of dermal fibroblasts and dermal papilla cells using nanovesicles extracted from stem cells using blue light-based photobiomodulation technology. Biomater Sci 2025; 13:1209-1221. [PMID: 39902823 DOI: 10.1039/d4bm01591f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Human dermal fibroblasts (hDFs) play a critical role in skin health by producing extracellular matrix (ECM) components essential for structural stability, while hair follicle dermal papilla cells (HFDPCs) are key to hair follicle growth and regeneration. However, factors such as UV radiation, oxidative stress, and aging impair the functions of hDFs and HFDPCs, leading to decrement in ECM production and skin maintenance and hair loss conditions like alopecia. Recent advances in nanovesicles (NVs) derived from human adipose-derived stem cells (hADSCs) have shown an innovative way in the regenerative medicine field, particularly with promise for enhancing the functionality of diverse cell types. NVs, filled with diverse bioactive molecules, are non-immunogenic, biologically stable, and capable of promoting cellular activities. To further enhance the therapeutic potential of NVs, photobiomodulation (PBM) using blue light has emerged as a promising application. Optimized blue light irradiation can induce moderate levels of reactive oxygen species production in hADSCs, activating signaling pathways that upregulate angiogenic and regenerative markers in hADSCs. In this study, blue light-irradiated NVs demonstrated superior efficacy in promoting hDF proliferation, ECM synthesis, and the functionality of HFDPCs, resulting in enhanced skin maintenance and hair follicle regeneration. This approach presents a safer and more efficient way for treating skin and hair disorders, highlighting the potential use of blue light-irradiated NVs as an innovative therapeutic strategy.
Collapse
Affiliation(s)
- Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Sang Yoon Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Jiseon An
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - You Bin Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
7
|
Liu FY, Cho YL, Fridayana FR, Niloofar L, Vo MN, Huang Y, Limanjaya A, Kwon MH, Ock J, Lee SJ, Yin GN, Lee NK, Ryu JK. MT-100, a human Tie2-agonistic antibody, improves penile neurovasculature in diabetic mice via the novel target Srpx2. Exp Mol Med 2025; 57:104-117. [PMID: 39741183 PMCID: PMC11799434 DOI: 10.1038/s12276-024-01373-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 09/05/2024] [Accepted: 09/30/2024] [Indexed: 01/02/2025] Open
Abstract
Diabetes is an incurable, chronic disease that can lead to many complications, including angiopathy, peripheral neuropathy, and erectile dysfunction (ED). The angiopoietin-Tie2 signaling pathway plays a critical role in blood vessel development, formation, remodeling, and peripheral nerve regeneration. Therefore, strategies for activating the Tie2 signaling pathway have been developed as potential therapies for neurovascular diseases. Here, we developed a human Tie2-agonistic antibody (MT-100) that not only resists Ang-2 antagonism and activates Tie2 signaling but also regulates a novel target, sushi repeat-containing protein X-linked 2 (Srpx2). This regulation led to the survival of vascular and neuronal cells, a reduction in the production of reactive oxygen species (ROS), activation of the PI3K/AKT/eNOS signaling pathway, increased expression of neurotrophic factors, and ultimately alleviation of ED in diabetic mice. Our findings not only provide conclusive evidence that MT-100 is a promising therapeutic strategy for the treatment of diabetic ED but also suggest it has substantial clinical applications for other complications associated with diabetes.
Collapse
Affiliation(s)
- Fang-Yuan Liu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Young-Lai Cho
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Fitri Rahma Fridayana
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Lashkari Niloofar
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Minh Nhat Vo
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Yan Huang
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea
| | - Anita Limanjaya
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Mi-Hye Kwon
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea
| | - Seon-Jin Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon, Republic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea.
| | - Nam-Kyung Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
- MabTics Co., Ltd., Daejeon, Republic of Korea.
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University College of Medicine, Incheon, Republic of Korea.
- Program in Biomedical Science & Engineering, Inha University, Incheon, Republic of Korea.
| |
Collapse
|
8
|
Chandimali N, Bak SG, Park EH, Lim HJ, Won YS, Kim EK, Park SI, Lee SJ. Free radicals and their impact on health and antioxidant defenses: a review. Cell Death Discov 2025; 11:19. [PMID: 39856066 PMCID: PMC11760946 DOI: 10.1038/s41420-024-02278-8] [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: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
Free radicals, characterized by the presence of unpaired electrons, are highly reactive species that play a significant role in human health. These molecules can be generated through various endogenous processes, such as mitochondrial respiration and immune cell activation, as well as exogenous sources, including radiation, pollution, and smoking. While free radicals are essential for certain physiological processes, such as cell signaling and immune defense, their overproduction can disrupt the delicate balance between oxidants and antioxidants, leading to oxidative stress. Oxidative stress results in the damage of critical biomolecules like DNA, proteins, and lipids, contributing to the pathogenesis of various diseases. Chronic conditions such as cancer, cardiovascular diseases, neurodegenerative disorders, and inflammatory diseases have been strongly associated with the harmful effects of free radicals. This review provides a comprehensive overview of the characteristics and types of free radicals, their mechanisms of formation, and biological impacts. Additionally, we explore natural compounds and extracts studied for their antioxidant properties, offering potential therapeutic avenues for managing free radical-induced damage. Future research directions are also discussed to advance our understanding and treatment of free radical-associated diseases.
Collapse
Affiliation(s)
- Nisansala Chandimali
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
- Applied Biological Engineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon, 34113, Korea
| | - Seon Gyeong Bak
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
| | - Eun Hyun Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Korea
| | - Hyung-Jin Lim
- Scripps Korea Antibody Institute, Chuncheon, 24341, Korea
| | - Yeong-Seon Won
- Division of Research Management, Department of Bioresource Industrialization, Honam National Institute of Biological Resource, Mokpo, 58762, Korea
| | - Eun-Kyung Kim
- Nutritional Education Major, Graduate School of Education, Dong-A University, Busan, 49315, Korea
| | - Sang-Ik Park
- Department of Veterinary Pathology, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Korea.
| | - Seung Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup, 56212, Korea.
- Applied Biological Engineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon, 34113, Korea.
| |
Collapse
|
9
|
Malhotra K, Malik A, Almalki WH, Sahebkar A, Kesharwani P. Reactive Oxygen Species and its Manipulation Strategies in Cancer Treatment. Curr Med Chem 2025; 32:55-73. [PMID: 37303173 DOI: 10.2174/0929867330666230609110455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023]
Abstract
Cancer is one of the serious diseases of modern times, occurring in all parts of the world and shows a wide range of effects on the human body. Reactive Oxygen Species (ROS) such as oxide and superoxide ions have both advantages and disadvantages during the progression of cancer, dependent on their concentration. It is a necessary part of the normal cellular mechanisms. Changes in its normal level can cause oncogenesis and other relatable problems. Metastasis can also be controlled by ROS levels in the tumor cells, which can be prevented by the use of antioxidants. However, ROS is also used for the initiation of apoptosis in cells by different mediators. There exists a cycle between the production of oxygen reactive species, their effect on the genes, role of mitochondria and the progression of tumors. ROS levels cause DNA damage by the oxidation process, gene damage, altered expression of the genes and signalling mechanisms. They finally lead to mitochondrial disability and mutations, resulting in cancer. This review summarizes the important role and activity of ROS in developing different types of cancers like cervical, gastric, bladder, liver, colorectal and ovarian cancers.
Collapse
Affiliation(s)
- Kabil Malhotra
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Arzoo Malik
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| |
Collapse
|
10
|
Keith WC, Hemmati F, Vaghasiya RS, Amiri F, Mistriotis P. Differential Effects of Confinement-Induced ROS Accumulation on Highly Motile Cancerous and Non-Cancerous Cells. AIChE J 2024; 70:e18598. [PMID: 40099227 PMCID: PMC11913314 DOI: 10.1002/aic.18598] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 08/20/2024] [Indexed: 03/19/2025]
Abstract
In vivo, migrating cells often encounter microenvironments that impose spatial constraints, leading to cell and nuclear deformation. As confinement-induced DNA damage has been linked to the accumulation of reactive oxygen species (ROS), we sought to investigate the impact of oxidative stress on cell behavior within confined spaces. Using microchannel devices that enable control of the degree and duration of cell confinement, we demonstrate that confined migration increases ROS levels in both HT-1080 fibrosarcoma cells and human dermal fibroblasts. Treatment with the antioxidant N-Acetyl-L-cysteine (NAC) counteracts confinement-induced ROS accumulation, suppressing p53 activation and supporting cell survival in both cell lines. This intervention preferentially reduces dorsal perinuclear actin fibers in confined cancer cells. Loss of these fibers is associated with reduced nuclear rupture frequency and increased confined migration. Collectively, this work provides insights into the differential effects of ROS on cancerous and non-cancerous cells and suggests that antioxidants may support tumor progression.
Collapse
Affiliation(s)
| | - Farnaz Hemmati
- Department of Chemical Engineering, Auburn University, Auburn AL, 36849, USA
| | | | - Farshad Amiri
- Department of Chemical Engineering, Auburn University, Auburn AL, 36849, USA
| | | |
Collapse
|
11
|
Amanda B, Pragasta R, Cakrasana H, Mustika A, Faizah Z, Oceandy D. The Hippo Signaling Pathway, Reactive Oxygen Species Production, and Oxidative Stress: A Two-Way Traffic Regulation. Cells 2024; 13:1868. [PMID: 39594616 PMCID: PMC11592687 DOI: 10.3390/cells13221868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The Hippo signaling pathway is recognized for its significant role in cell differentiation, proliferation, survival, and tissue regeneration. Recently, the Hippo signaling pathway was also found to be associated with oxidative stress and reactive oxygen species (ROS) regulation, which are important in the regulation of cell survival. Studies indicate a correlation between components of the Hippo signaling pathway, including MST1, YAP, and TAZ, and the generation of ROS. On the other hand, ROS and oxidative stress can activate key components of the Hippo signaling pathway. For example, ROS production activates MST1, which subsequently phosphorylates FOXO3, leading to apoptotic cell death. ROS was also found to regulate YAP, in addition to MST1/2. Oxidative stress and ROS formation can impair lipids, proteins, and DNA, leading to many disorders, including aging, neurodegeneration, atherosclerosis, and diabetes. Consequently, understanding the interplay between the Hippo signaling pathway, ROS, and oxidative stress is crucial for developing future disease management strategies. This paper aimed to review the association between the Hippo signaling pathway, regulation of ROS production, and oxidative stress to provide beneficial information in understanding cell function and pathological processes.
Collapse
Affiliation(s)
- Bella Amanda
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (R.P.); (H.C.); (Z.F.)
- Airlangga University Teaching Hospital, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Rangga Pragasta
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (R.P.); (H.C.); (Z.F.)
- Faculty of Medicine, Universitas Islam Malang, Malang 65144, Indonesia
| | - Haris Cakrasana
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (R.P.); (H.C.); (Z.F.)
| | - Arifa Mustika
- Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia;
| | - Zakiyatul Faizah
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (R.P.); (H.C.); (Z.F.)
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK;
| |
Collapse
|
12
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [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: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
13
|
Azevedo T, Ferreira T, Peña‐Corona SI, Cortes H, Silva‐Reis R, da Costa RMG, Faustino‐Rocha AI, Oliveira PA, Calina D, Cardoso SM, Büsselberg D, Leyva‐Gómez G, Sharifi‐Rad J, Cho WC. Natural products‐based antiangiogenic agents: New frontiers in cancer therapy. FOOD FRONTIERS 2024; 5:2423-2466. [DOI: 10.1002/fft2.466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
AbstractAngiogenesis, vital for tumor growth and metastasis, is a promising target in cancer therapy. Natural compounds offer potential as antiangiogenic agents with reduced toxicity. This review provides a comprehensive overview of natural product‐based antiangiogenic therapies, focusing on molecular mechanisms and therapeutic potential. A systematic search identified relevant articles from 2019 to 2023. Various natural compounds, including polyphenols, terpenes, alkaloids, cannabinoids, omega‐3 fatty acids, polysaccharides, proteins, and carotenoids, were investigated for their antiangiogenic properties. Challenges such as dose standardization, routes of administration, and potential side effects remain. Further studies, including in‐depth animal models and human epidemiological studies, must elucidate clinical efficacy and safety. Synergistic effects with current antiangiogenic therapies, such as bevacizumab and tyrosine kinase inhibitors, should be explored. Additionally, the potential hormone‐dependent effects of compounds like genistein highlight the need for safety evaluation. In conclusion, natural products hold promise as adjunctive therapies to conventional antineoplastic drugs in modulating angiogenesis in cancer. However, robust clinical trials are needed to validate preclinical findings and ensure safety and efficacy.
Collapse
Affiliation(s)
- Tiago Azevedo
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Tiago Ferreira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Sheila I. Peña‐Corona
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Hernán Cortes
- Laboratorio de Medicina Genómica, Departamento de Genómica Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra Ciudad de México Mexico
| | - Rita Silva‐Reis
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | - Rui M. Gil da Costa
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI‐IPOP)/RISE@CI‐IPOP (Health Research Network) Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto. CCC) Porto Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering University of Porto Porto Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering University of Porto Porto Portugal
- Postgraduate Programme in Adult Health (PPGSAD), Department of Morphology Federal University of Maranhão (UFMA), UFMA University Hospital (HUUFMA) São Luís Brazil
| | - Ana I. Faustino‐Rocha
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
- Comprehensive Health Research Center, Department of Zootechnics, School of Sciences and Technology University of Évora Evora Portugal
| | - Paula A. Oliveira
- Centre for the Research and Technology of Agro‐Environmental and Biological Sciences (CITAB), Inov4Agro University of Trás‐os‐Montes and Alto Douro (UTAD) Vila Real Portugal
| | - Daniela Calina
- Department of Clinical Pharmacy University of Medicine and Pharmacy of Craiova Craiova Romania
| | - Susana M. Cardoso
- LAQV‐REQUIMTE, Department of Chemistry University of Aveiro Aveiro Portugal
| | | | - Gerardo Leyva‐Gómez
- Departamento de Farmacia, Facultad de Química Universidad Nacional Autónoma de México Ciudad de México Mexico
| | - Javad Sharifi‐Rad
- Centro de Estudios Tecnológicos y Universitarios del Golfo Veracruz Mexico
- Department of Medicine, College of Medicine Korea University Seoul Republic of Korea
- Facultad de Medicina Universidad del Azuay Cuenca Ecuador
| | - William C. Cho
- Department of Clinical Oncology Queen Elizabeth Hospital Kowloon Hong Kong
| |
Collapse
|
14
|
Zhou W, Xu C, Niu J, Xiong Y, He Z, Xu H, Zhang M, Wang H, Xu Q, Wang X, Wang Z. Inhibitory effects of Eplerenone on angiogenesis via modulating SGK1/TGF-β pathway in contralateral kidney of CKD pregnancy rats. Cell Signal 2024; 122:111346. [PMID: 39147296 DOI: 10.1016/j.cellsig.2024.111346] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Eplerenone is a selective aldosterone receptor blocker that is effective in preventing the progression of chroinic kidney disease (CKD). However, its mechanism and role in CKD pregnancy still remain uncertain. The aim of this study was to evaluate whether eplerenone could attenuated the fibrosis of unilateral ureteral obstruction (UUO) pregnant rats' contralateral kidney, improved pregnancy outcome and explore its therapeutic mechanisms. METHODS A pregnancy rat model of UUO established, female Wistar rats were randomly assigned into sham-operated group (Sham group),sham-operated combined pregnancy group (SP group), unilateral ureteral obstruction combined pregnancy group (UUO + Pregnancy group), unilateral ureteral obstruction combined pregnancy, administered eplerenone (UUO + Pregnancy+Eplerenone group). On the 18th day of pregnancy, the rats were placed in a metabolic cage, 24 h urine was collected and stored at -80 °C. Next day, all animals were euthanized, and serum was collected by centrifugation and stored at -20 °C. Then the right kidney was extracted, a part of the kidney was placed in 4% paraformaldehyde for morphology, immunohistochemical staining, and immunofluorescence staining, and the other part was placed in a - 80 °C refrigerator for RNA and protein extraction. In vitro, HUVECs was treated with aldosterone, progesterone and estradiol, VEGFA and its receptor blocker bevacizumab. The ability of proliferation, migration and tubularization of HUVECs was detected by CCK-8, scratch wound assay and endothelial tube formation assay. And the co-expression of CD34 and α-SMA of HUVECs was detected by Flow cytometry. RESULTS Immunofluorescence results showed that the co-expression of CD34 and α-SMA increased in the UUO + Pregnancy group was significantly increased. The expression of SGK-1, TGFβ-1, Smad2, Smad3, VEGF-A, VEGFR2, CD34, α-SMA and Collagen I was significantly higher in the kidneys of the UUO + Pregnancy group compared to the Sham group and SP group. Eplerenone inhibited the expression of those results. In vitro, the ability of proliferation, migration and tubularization was increased after treated with aldosterone, aldosterone with progesterone and estradiol or VEGFA. Similarly, the expression of α-SMA on the surface of HUVECs treated with aldosterone, aldosterone with progesterone and estradiol were increased, while eplerenone supressed its expression. CONCLUSION Eplerenone inhibits renal angiogenesis by blocking the SGK-1/TGFβ signal transduction pathway, thereby inhibiting the phenotypic transformation of endothelial cells, slowing down renal fibrosis, and reducing kidney damage caused by pregnancy.
Collapse
Affiliation(s)
- Wenping Zhou
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Chang Xu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Jieqi Niu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yunzhao Xiong
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Zhen He
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hepeng Xu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Mengjuan Zhang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Hongshuang Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Qingyou Xu
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Xiangting Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
| | - Zheng Wang
- Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Hebei University of Chinese Medicine, Shijiazhuang, China; Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, China; College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China.
| |
Collapse
|
15
|
Fiehn LA, Kunisch E, Saur M, Arango-Ospina M, Merle C, Hagmann S, Stiller A, Hupa L, Kaňková H, Galusková D, Renkawitz T, Boccaccini AR, Westhauser F. A comparative in vitro and in vivo analysis of the impact of copper substitution on the cytocompatibility, osteogenic, and angiogenic properties of a borosilicate bioactive glass. J Biomed Mater Res A 2024; 112:1740-1759. [PMID: 38623001 DOI: 10.1002/jbm.a.37721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/10/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024]
Abstract
The 0106-B1-bioactive glass (BG) composition (in wt %: 37.5 SiO2, 22.6 CaO, 5.9 Na2O, 4.0 P2O5, 12.0 K2O, 5.5 MgO, and 12.5 B2O3) has demonstrated favorable processing properties and promising bone regeneration potential. The present study aimed to evaluate the biological effects of the incorporation of highly pro-angiogenic copper (Cu) in 0106-B1-BG in vitro using human bone marrow-derived mesenchymal stromal cells (BMSCs) as well as its in vivo potential for bone regeneration. CuO was added to 0106-B1-BG in exchange for CaO, resulting in Cu-doped BG compositions containing 1.0, 2.5 and 5.0 wt % CuO (composition in wt %: 37.5 SiO2, 21.6/ 20.1/17.6 CaO, 5.9 Na2O, 4.0 P2O5, 12.0 K2O, 5.5 MgO, 12.5 B2O3, and 1.0/ 2.5/ 5.0 CuO). In vitro, the BGs' impact on the viability, proliferation, and growth patterns of BMSCs was evaluated. Analyses of protein secretion, matrix formation, and gene expression were used for the assessment of the BGs' influence on BMSCs regarding osteogenic differentiation and angiogenic stimulation. The presence of Cu improved cytocompatibility, osteogenic differentiation, and angiogenic response when compared with unmodified 0106-B1-BG in vitro. In vivo, a critical-size femoral defect in rats was filled with scaffolds made from BGs. Bone regeneration was evaluated by micro-computed tomography. Histological analysis was performed to assess bone maturation and angiogenesis. In vivo effects regarding defect closure, presence of osteoclastic cells or vascular structures in the defect were not significantly changed by the addition of Cu compared with undoped 0106-B1-BG scaffolds. Hence, while the in vitro properties of the 0106-B1-BG were significantly improved by the incorporation of Cu, further evaluation of the BG composition is necessary to transfer these effects to an in vivo setting.
Collapse
Affiliation(s)
- Linn Anna Fiehn
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Elke Kunisch
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Merve Saur
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Christian Merle
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Sébastien Hagmann
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Adrian Stiller
- Johan Gadolin Process Chemistry Centre, Åbo Akademi University, Turku, Finland
| | - Leena Hupa
- Johan Gadolin Process Chemistry Centre, Åbo Akademi University, Turku, Finland
| | - Hana Kaňková
- Centre for Functional and Surface Functionalized Glass, Alexander Dubček University of Trenčín, Trenčín, Slovakia
| | - Dagmar Galusková
- Centre for Functional and Surface Functionalized Glass, Alexander Dubček University of Trenčín, Trenčín, Slovakia
| | - Tobias Renkawitz
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Fabian Westhauser
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
16
|
Shin S, Kim MH, Oh CM, Ha E, Ryoo JH. Association Between Type 1 Diabetes Mellitus and Incident Gastrointestinal Cancer in Korean Population: A Nationwide Retrospective Cohort Study. Diabetes Metab Res Rev 2024; 40:e3848. [PMID: 39376128 DOI: 10.1002/dmrr.3848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/11/2024] [Accepted: 09/13/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND The age-standardised incidence ratio of gastrointestinal cancers in type 1 diabetes (T1D) patients has been reported to be higher than that in the general population. After adjusting for shared risk factors, we aimed to explore the association between T1D and gastrointestinal cancer and examine how this relationship varies by age and sex. MATERIALS AND METHODS This retrospective cohort study included 268,179 participants from the Korean National Health Insurance Service-National Sample Cohort. The primary outcome is the incident of gastrointestinal cancers, based on diagnostic codes. Multivariate Cox regression analyses were performed to assess the association between T1D and gastrointestinal cancers. RESULTS Of the 268,179 participants, 2681 had T1D at baseline and were followed for 12.98 (± 2.92) years. Compared with non-T1D, T1D patients had a significantly increased risk of all gastrointestinal cancer (adjusted hazard ratio [aHR]: 1.403, 95% confidence interval [CI]: 1.253-1.573). T1D patients increased risks of oesophageal cancer (aHR: 1.864, 95% CI: 1.038-3.349), gastric cancer (aHR: 1.313, 95% CI: 1.066-1.616), colon cancer (aHR: 1.365, 95% CI: 1.110-1.678), liver cancer (aHR: 1.388, 95% CI: 1.115-1.727), and pancreatic cancer (aHR: 1.716, 95% CI: 1.182-2.492). The consistency of this association persisted among both male and female, with its strength increasing with older age. CONCLUSIONS The risk of gastrointestinal cancer was significantly increased in T1D patients. Older male T1D patients exhibit a higher risk, suggesting the need for targeted attention in their care.
Collapse
Affiliation(s)
- Soonsu Shin
- Department of Preventive Medicine, Graduate School, Kyung Hee University, Seoul, Korea
- Department of Occupational and Environmental Medicine, Kyung Hee University Hospital, Seoul, Korea
| | - Min-Ho Kim
- Ewha Medical Data Organization, Ewha Womans University Seoul Hospital, Seoul, Korea
| | - Chang-Mo Oh
- Department of Preventive Medicine, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Eunhee Ha
- Department of Occupational and Environment Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Jae-Hong Ryoo
- Department of Occupational and Environmental Medicine, School of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
17
|
Song J, Feng Y, Yan J, Wang Y, Yan W, Yang N, Wu T, Liu S, Wang Y, Zheng N, He L, Zhang Y. Computed Tomography Imaging Guided Microenvironment-Responsive Ir@WO 3-x Dual-Catalytic Nanoreactor for Selective Radiosensitization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405192. [PMID: 39102342 PMCID: PMC11481196 DOI: 10.1002/advs.202405192] [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: 05/13/2024] [Revised: 07/28/2024] [Indexed: 08/07/2024]
Abstract
Radiotherapy (RT) is often administered, either alone or in combination with other therapies, for most malignancies. However, the degree of tumor oxygenation, damage to adjacent healthy tissues, and inaccurate guidance remain issues that result in discontinuation or failure of RT. Here, a multifunctional therapeutic platform based on Ir@WO3-x is developed which simultaneously addresses these critical issues above for precision radiosensitization. Ir@WO3-x nanoreactors exhibit strong absorption of X-ray, acting as radiosensitizers. Moreover, ultrasmall Ir enzyme-mimic nanocrystals (NCs) are decorated onto the surface of the nanoreactor, where NCs have catalyst-like activity and are sensitive to H2O2 in the tumor microenvironment (TME) under near infrared-II (NIR-II) light stimulation. They efficiently catalyze the conversion of H2O2 to O2, thereby ameliorating hypoxia, inhibiting the expression of HIF-1α, and enhancing RT-induced DNA damage in cancerous tissue, further improving the efficiency of RT. Additionally, in response to high H2O2 levels in TME, the Ir@WO3-x nanoreactor also exerts peroxidase-like activity, boosting exogenous ROS, which increases oxidative damage and enhances ROS-dependent death signaling. Furthermore, Ir@WO3-x can serve as a high-quality computed tomography contrast agent due to its high X-ray attenuation coefficient and generation of pronounced tumor-tissue contrast. This report highlights the potential of advanced health materials to enhance precision therapeutic modalities.
Collapse
Affiliation(s)
- Jiayu Song
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
| | - Yue Feng
- Department of Gynecological OncologyZhejiang Cancer HospitalZhengzhouZhejiang310022China
| | - Jiazhuo Yan
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Ying Wang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Weixiao Yan
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
| | - Nan Yang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Tusheng Wu
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Sijia Liu
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Yuan Wang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| | - Nannan Zheng
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhouHenan450000China
| | - Liangcan He
- School of Medicine and HealthKey Laboratory of Microsystems and Microstructures ManufacturingHarbin Institute of TechnologyHarbin150001China
- Zhengzhou Research InstituteHarbin Institute of TechnologyZhengzhouHenan450000China
| | - Yunyan Zhang
- Department of Gynecological RadiotherapyHarbin Medical University Cancer HospitalHarbin150001China
| |
Collapse
|
18
|
Kourdova LT, Miranda AL, Ovejero M, Anastasía A, Genti-Raimondi S, Racca AC, Panzetta-Dutari GM. Krüppel-like factor 6 involvement in the endoplasmic reticulum homeostasis of extravillous trophoblasts. Placenta 2024; 155:42-51. [PMID: 39121586 DOI: 10.1016/j.placenta.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION Trophoblast homeostasis and differentiation require a proper endoplasmic reticulum (ER) function. The Krüppel-like factor-6 (KLF6) transcription factor modulates trophoblast migration, differentiation, and reactive oxygen species (ROS) production. Since ROS may impact on ER homeostasis, we assessed whether downregulation of KLF6 altered the unfolded protein response (UPR) and cellular process associated with ER homeostasis. MATERIALS AND METHODS Protein and RNA expression were analyzed by Western blot and qRT-PCR, respectively, in extravillous trophoblast HTR-8/SVneo cells silenced for KLF6. Apoptosis was detected by flow cell cytometry using Annexin V Apoptosis Detection Kit. Protein trafficking was assessed by confocal microscopy of a reporter fluorescent protein whose release from the ER was synchronized. RESULTS KLF6 downregulation reduced the expression of BiP, the master regulator of the UPR, at protein, mRNA, and pre-mRNA levels. Ire1α protein, XBP1 splicing, and DNAJB9 mRNA levels were also reduced in KLF6-silenced cells. Instead, PDI, Ero1α, and the p-eIF2α/eIF2α ratio as well as autophagy and proteasome dependent protein degradation remained unchanged while intracellular trafficking was increased. Under thapsigargin-induced stress, KLF6 silencing impaired BiP protein and mRNA expression increase, as well as the activation of the Ire1α pathway, but it raised the p-eIF2α/eIF2α ratio and CHOP protein levels. Nevertheless, apoptosis was not increased. DISCUSSION Results provide the first evidence of KLF6 as a modulator of the UPR components. The increase in protein trafficking and protection from apoptosis, observed in KLF6-silenced cells, are consistent with its role in extravillous trophoblast migration and differentiation.
Collapse
Affiliation(s)
- Lucille T Kourdova
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Andrea L Miranda
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Milagros Ovejero
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Agustín Anastasía
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Susana Genti-Raimondi
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Ana C Racca
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina
| | - Graciela M Panzetta-Dutari
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Bioquímica Clínica, Ciudad Universitaria, X5000HUA, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Ciudad Universitaria, X5000HUA, Córdoba, Argentina.
| |
Collapse
|
19
|
Di Carlo E, Sorrentino C. Oxidative Stress and Age-Related Tumors. Antioxidants (Basel) 2024; 13:1109. [PMID: 39334768 PMCID: PMC11428699 DOI: 10.3390/antiox13091109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Oxidative stress is the result of the imbalance between reactive oxygen and nitrogen species (RONS), which are produced by several endogenous and exogenous processes, and antioxidant defenses consisting of exogenous and endogenous molecules that protect biological systems from free radical toxicity. Oxidative stress is a major factor in the aging process, contributing to the accumulation of cellular damage over time. Oxidative damage to cellular biomolecules, leads to DNA alterations, lipid peroxidation, protein oxidation, and mitochondrial dysfunction resulting in cellular senescence, immune system and tissue dysfunctions, and increased susceptibility to age-related pathologies, such as inflammatory disorders, cardiovascular and neurodegenerative diseases, diabetes, and cancer. Oxidative stress-driven DNA damage and mutations, or methylation and histone modification, which alter gene expression, are key determinants of tumor initiation, angiogenesis, metastasis, and therapy resistance. Accumulation of genetic and epigenetic damage, to which oxidative stress contributes, eventually leads to unrestrained cell proliferation, the inhibition of cell differentiation, and the evasion of cell death, providing favorable conditions for tumorigenesis. Colorectal, breast, lung, prostate, and skin cancers are the most frequent aging-associated malignancies, and oxidative stress is implicated in their pathogenesis and biological behavior. Our aim is to shed light on the molecular and cellular mechanisms that link oxidative stress, aging, and cancers, highlighting the impact of both RONS and antioxidants, provided by diet and exercise, on cellular senescence, immunity, and development of an antitumor response. The dual role of ROS as physiological regulators of cell signaling responsible for cell damage and diseases, as well as its use for anti-tumor therapeutic purposes, will also be discussed. Managing oxidative stress is crucial for promoting healthy aging and reducing the risk of age-related tumors.
Collapse
Affiliation(s)
- Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
20
|
Guo L, Ding Z, Hu J, Liu S. Efficient Encapsulation of β-Lapachone into Self-Immolative Polymer Nanoparticles for Cyclic Amplification of Intracellular Reactive Oxygen Species Stress. ACS NANO 2024. [PMID: 39263977 DOI: 10.1021/acsnano.4c09232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
The selective upregulation of intracellular oxidative stress in cancer cells presents a promising approach for effective cancer treatment. In this study, we report the integration of enzyme catalytic amplification and chemical amplification reactions in β-lapachone (Lap)-loaded micellar nanoparticles (NPs), which are self-assembled from reactive oxygen species (ROS)-responsive self-immolative polymers (SIPs). This integration enables cyclic amplification of intracellular oxidative stress in cancer cells. Specifically, we have developed ROS-responsive SIPs with phenylboronic ester triggering motifs and hexafluoroisopropanol moieties in the side chains, significantly enhancing Lap loading efficiency (98%) and loading capacity (33%) through multiple noncovalent interactions. Upon ROS activation in tumor cells, the Lap-loaded micellar NPs disassemble, releasing Lap and generating additional ROS via enzyme catalytic amplification. This process elevates intracellular oxidative stress and triggers polymer depolymerization in a positive feedback loop. Furthermore, the degradation of SIPs via chemical amplification produces azaquinone methide intermediates, which consume intracellular thiol-related substrates, disrupt intracellular redox hemostasis, further intensify oxidative stress, and promote cancer cell apoptosis. This work introduces a strategy to enhance intracellular oxidative stress by combining enzymatic and chemical amplification reactions, providing a potential pathway for the development of highly efficient anticancer agents.
Collapse
Affiliation(s)
- Lingxiao Guo
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| | - Zexuan Ding
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| | - Jinming Hu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| | - Shiyong Liu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province 230026, China
| |
Collapse
|
21
|
Nadigar S, Gattu R, Ramesh S, Dharmappa RN, Nanjundaswamy VK, Ramesh S. A novel class of potent antiangiogenic and antioxidant pyrazoles: synthesis, bioactivity, docking and ADMET studies. Future Med Chem 2024; 16:2285-2300. [PMID: 39263822 PMCID: PMC11622771 DOI: 10.1080/17568919.2024.2394020] [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: 06/19/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
Aim: Angiogenesis is the hallmark of cancer progression driven by VEGF/VEGFR-2 signalling pathway, inhibition of which could be a solution to tackle the progression of tumour cells and thus arresting their growth.Materials & methods: A novel class of pyrazoles was synthesized using arginine and dibromo ketones. Antiangiogenic activity was performed by in vivo yolk sac method. Antioxidant activity was evaluated by hydroxyl and superoxide radical scavenging assays. Docking studies were performed to determine the pyrazoles' binding potential with VEGFR-2 receptor and VEGF tyrosine kinase. ADMET properties were calculated using SwissADME and admetSAR for drug-likeness.Results: Compounds 5a-e showed significant antiangiogenic effects. Compound 5f exhibited effective hydroxyl and superoxide radical scavenging activities. Docking results confirmed the potential binding efficiency with VEGFR-2 receptor over VEGF tyrosine kinase, thus, functioning as competitive-inhibitors. ADMET studies revealed that the compounds possess favourable drug-like qualities.Conclusion: This study presents a novel class of pyrazoles as promising antioxidant and antiangiogenic agents with favourable drug-likeness properties.
Collapse
Affiliation(s)
- Siddaram Nadigar
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce & Science (a recognized Research Centre of University of Mysore), Ooty Road, Mysuru-25, Karnataka, India
| | - Rohith Gattu
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce & Science (a recognized Research Centre of University of Mysore), Ooty Road, Mysuru-25, Karnataka, India
| | - Sanjay Ramesh
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce & Science (a recognized Research Centre of University of Mysore), Ooty Road, Mysuru-25, Karnataka, India
| | - Rekha N Dharmappa
- Postgraduate Department of Biotechnology, JSS College of Arts, Commerce & Science (a recognized Research Centre of University of Mysore), Ooty Road, Mysuru-25, Karnataka, India
| | - Vijendra Kumar Nanjundaswamy
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce & Science (a recognized Research Centre of University of Mysore), Ooty Road, Mysuru-25, Karnataka, India
| | - Suhas Ramesh
- Postgraduate Department of Chemistry, JSS College of Arts, Commerce & Science (a recognized Research Centre of University of Mysore), Ooty Road, Mysuru-25, Karnataka, India
| |
Collapse
|
22
|
Panda B, Tripathy A, Patra S, Kullu B, Tabrez S, Jena M. Imperative connotation of SODs in cancer: Emerging targets and multifactorial role of action. IUBMB Life 2024; 76:592-613. [PMID: 38600696 DOI: 10.1002/iub.2821] [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: 01/08/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024]
Abstract
Superoxide dismutase (SOD) is a crucial enzyme responsible for the redox homeostasis inside the cell. As a part of the antioxidant defense system, it plays a pivotal role in the dismutation of the superoxide radicals (O 2 - ) generated mainly by the oxidative phosphorylation, which would otherwise bring out the redox dysregulation, leading to higher reactive oxygen species (ROS) generation and, ultimately, cell transformation, and malignancy. Several studies have shown the involvement of ROS in a wide range of human cancers. As SOD is the key enzyme in regulating ROS, any change, such as a transcriptional change, epigenetic remodeling, functional alteration, and so forth, either activates the proto-oncogenes or aberrant signaling cascades, which results in cancer. Interestingly, in some cases, SODs act as tumor promoters instead of suppressors. Furthermore, SODs have also been known to switch their role during tumor progression. In this review, we have tried to give a comprehensive account of SODs multifactorial role in various human cancers so that SODs-based therapeutic strategies could be made to thwart cancers.
Collapse
Affiliation(s)
- Biswajit Panda
- Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Ankita Tripathy
- Post Graduate Department of Botany, Utkal University, Bhubaneswar, India
| | - Srimanta Patra
- Post Graduate Department of Botany, Berhampur University, Berhampur, India
| | - Bandana Kullu
- Post Graduate Department of Botany, Utkal University, Bhubaneswar, India
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mrutyunjay Jena
- Post Graduate Department of Botany, Berhampur University, Berhampur, India
| |
Collapse
|
23
|
Vornic I, Nesiu A, Ardelean AM, Todut OC, Pasare VC, Onel C, Raducan ID, Furau CG. Antioxidant Defenses, Oxidative Stress Responses, and Apoptosis Modulation in Spontaneous Abortion: An Immunohistochemistry Analysis of First-Trimester Chorionic Villi. Life (Basel) 2024; 14:1074. [PMID: 39337859 PMCID: PMC11432807 DOI: 10.3390/life14091074] [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: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Oxidative stress (OS) and apoptosis are critical factors in placental development and function. Their interplay influences trophoblast proliferation, differentiation, and invasion, as well as vascular development. An imbalance between these processes can lead to pregnancy-related disorders such as preeclampsia, intrauterine growth restriction, and even spontaneous abortion. Our study seeks to elucidate the associations between preventive antioxidant/protective OS response factors-glutathione (GSH), MutT Homolog 1 (MTH1), and apoptotic regulation modulators-tumor protein p53 and B-cell lymphoma (Bcl-2) transcripts, in the context of spontaneous abortion (30 samples) versus elective termination of pregnancy (20 samples), using immunohistochemistry (IHC) to determine their proteomic expression in chorionic villi within abortive fetal placenta tissue samples. Herein, comparative statistical analyses revealed that both OS response factors, GSH and MTH1, were significantly under-expressed in spontaneous abortion cases as compared to elective. Conversely, for apoptotic regulators, p53 expression was significantly higher in spontaneous abortion cases, whereas Bcl-2 expression was significantly lower in spontaneous abortion cases. These findings suggest that a strong pro-apoptotic signal is prevalent within spontaneous abortion samples, alongside reduced anti-apoptotic protection, depleted antioxidant defenses and compromised oxidative DNA damage prevention/repair, as compared to elective abortion controls. Herein, our hypothesis that OS and apoptosis are closely linked processes contributing to placental dysfunction and spontaneous abortion was thus seemingly corroborated. Our results further highlight the importance of maintaining redox homeostasis and apoptotic regulation for a successful pregnancy. Understanding the mechanisms underlying this interplay is essential for developing potential therapies to manage OS, promote placentation, and avoid unwanted apoptosis, ultimately improving pregnancy outcomes. Antioxidant supplementation, modulation of p53 activity, and the enhancement of DNA repair mechanisms may represent potential approaches to mitigate OS and apoptosis in the placenta. Further research is needed to explore these strategies and their efficacy in preventing spontaneous abortion.
Collapse
Affiliation(s)
- Ioana Vornic
- Doctoral School, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
- Discipline of Gynecology, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
| | - Alexandru Nesiu
- Discipline of Urology, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
| | - Ana Maria Ardelean
- Doctoral School, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
- Discipline of Gynecology, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
| | - Oana Cristina Todut
- Doctoral School, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
- Discipline of Gynecology, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
| | - Victoria Cristina Pasare
- Doctoral School, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
- Discipline of Gynecology, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
| | - Cristina Onel
- Doctoral School, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
- Discipline of Gynecology, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
| | - Ionuț Daniel Raducan
- Doctoral School, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
| | - Cristian George Furau
- Discipline of Gynecology, Faculty of Medicine, "Vasile Goldiș" Western University of Arad, Liviu Rebreanu Street, No. 86, 310414 Arad, Romania
| |
Collapse
|
24
|
Wang L, Qiu N, Tong S, Yu Y, Xi S, Wang F. Matrine Suppresses Arsenic-Induced Malignant Transformation of SV-HUC-1 Cells via NOX2. Int J Mol Sci 2024; 25:8878. [PMID: 39201564 PMCID: PMC11354282 DOI: 10.3390/ijms25168878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Arsenic (As) has been classified as a carcinogen for humans. There is abundant evidence indicating that arsenic increases the risk of bladder cancer among human populations. However, the underlying mechanisms have yet to be fully understood and elucidated. NADPH oxidases (NOXs) are the main enzymes for ROS production in the body. NADPH Oxidase 2 (NOX2), which is the most distinctive and ubiquitously expressed subunit of NOXs, can promote the formation and development of tumors. The utilization of NOX2 as a therapeutic target has been proposed to modulate diseases resulting from the activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3). Matrine has been reported to exhibit various pharmacological effects, including anti-inflammatory, antifibrotic, antitumor, and analgesic properties. However, it has not been reported whether matrine can inhibit malignant transformation induced by arsenic in uroepithelial cells through NOX2. We have conducted a series of experiments using both a sub-chronic NaAsO2 exposure rat model and a long-term NaAsO2 exposure cell model. Our findings indicate that arsenic significantly increases cell proliferation, migration, and angiogenesis in vivo and in vitro. Arsenic exposure resulted in an upregulation of reactive oxygen species (ROS), NOX2, and NLRP3 inflammasome expression. Remarkably, both in vivo and in vitro, the administration of matrine demonstrated a significant improvement in the detrimental impact of arsenic on bladder epithelial cells. This was evidenced by the downregulation of proliferation, migration, and angiogenesis, as well as the expression of the NOX2 and NLRP3 inflammasomes. Collectively, these findings indicate that matrine possesses the ability to reduce NOX2 levels and inhibit the transformation of bladder epithelial cells.
Collapse
Affiliation(s)
- Lanfei Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Nianfeng Qiu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Suyuan Tong
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Yan Yu
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Shuhua Xi
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| | - Fei Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang 110122, China
- Department of Environmental Health, School of Public Health, China Medical University, Shenyang 110122, China
- Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, Shenyang 110122, China
| |
Collapse
|
25
|
Hegde M, Girisa S, Devanarayanan TN, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara AB. Network of Extracellular Traps in the Pathogenesis of Sterile Chronic Inflammatory Diseases: Role of Oxidative Stress and Potential Clinical Applications. Antioxid Redox Signal 2024; 41:396-427. [PMID: 37725535 DOI: 10.1089/ars.2023.0329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Thulasidharan Nair Devanarayanan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Mohammed S Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- BioImaging Unit, Space Research Centre, Michael Atiyah Building, University of Leicester, Leicester, United Kingdom
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
26
|
Ollila E, Ahtikoski A, Puistola U, Karihtala P, Urpilainen E. Redox-state-regulating enzymes have prognostic value in diabetic endometrial cancer patients: impact of statin use? Front Oncol 2024; 14:1393103. [PMID: 39040459 PMCID: PMC11260677 DOI: 10.3389/fonc.2024.1393103] [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: 02/28/2024] [Accepted: 05/30/2024] [Indexed: 07/24/2024] Open
Abstract
Objective The incidence of endometrial cancer is increasing, and oxidative stress has been suggested to play a vital role in its carcinogenesis. Statins have an impact on the cellular redox-state. The aim of this study was to determine the effects of statin use on redox-state regulating enzymes in endometrial cancer in women with type 2 diabetes. Materials and methods This retrospective study consisted of 119 women with type 2 diabetes who were diagnosed with endometrial cancer at Oulu University Hospital in Finland between 2007 and 2014. There were 58 statin users and 61 non-users based on medication use at the time of endometrial cancer diagnosis. The expression of redox-state regulating proteins nuclear factor erythroid 2-related factor 2 (Nrf2) and Kelch-like ECH-associated protein 1 (Keap1) in the tumor samples was assessed immunohistochemically, and manganese superoxide dismutase (MnSOD) levels were measured both immunohistochemically and from serum samples. Results High MnSOD expression predicted better progression-free survival (PFS) in statin non-users in a univariate analysis (p=0.02). There was no statistical difference in overall survival (OS) or PFS between strong and weak expression of Nrf2 and Keap1. After adjusting for stage and statin use, the results were similar. Conclusion Statin non-users with strong MnSOD expression had better PFS compared to statin users which proves that statins have impact on redox-state regulating enzymes. However, these findings are preliminary and require further research.
Collapse
Affiliation(s)
- Elina Ollila
- Department of Obstetrics and Gynecology, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, and University of Oulu, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, University of Oulu, Oulu, Finland
| | - Anne Ahtikoski
- Department of Pathology, Fimlab Laboratories, University of Tampere, Tampere, Finland
| | - Ulla Puistola
- Department of Obstetrics and Gynecology, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, and University of Oulu, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, University of Oulu, Oulu, Finland
| | - Peeter Karihtala
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, Helsinki, Finland
| | - Elina Urpilainen
- Department of Obstetrics and Gynecology, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, and University of Oulu, Oulu, Finland
- Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
- Medical Research Center, Oulu University Hospital, Wellbeing Services County of North Ostrobothnia, University of Oulu, Oulu, Finland
| |
Collapse
|
27
|
Hou Y, Wang H, Wu J, Guo H, Chen X. Dissecting the pleiotropic roles of reactive oxygen species (ROS) in lung cancer: From carcinogenesis toward therapy. Med Res Rev 2024; 44:1566-1595. [PMID: 38284170 DOI: 10.1002/med.22018] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 01/30/2024]
Abstract
Lung cancer is a major cause of morbidity and mortality. The specific pulmonary structure to directly connect with ambient air makes it more susceptible to damage from airborne toxins. External oxidative stimuli and endogenous reactive oxygen species (ROS) play a crucial role in promoting lung carcinogenesis and development. The biological properties of higher ROS levels in tumor cells than in normal cells make them more sensitive and vulnerable to ROS injury. Therefore, the strategy of targeting ROS has been proposed for cancer therapy for decades. However, it is embarrassing that countless attempts at ROS-based therapies have had very limited success, and no FDA approval in the anticancer list was mechanistically based on ROS manipulation. Even compared with the untargetable proteins, such as transcription factors, ROS are more difficult to be targeted due to their chemical properties. Thus, the pleiotropic roles of ROS provide therapeutic potential for anticancer drug discovery, while a better dissection of the mechanistic action and signaling pathways is a prerequisite for future breakthroughs. This review discusses the critical roles of ROS in cancer carcinogenesis, ROS-inspired signaling pathways, and ROS-based treatment, exemplified by lung cancer. In particular, an eight considerations rule is proposed for ROS-targeting strategies and drug design and development.
Collapse
Affiliation(s)
- Ying Hou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Heng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, China
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Key Laboratory of Research and Evaluation of Bioactive Molecules & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China
- Department of Pharmaceutical Sciences, University of Macau, Taipa, Macao, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao, China
| |
Collapse
|
28
|
Wu B, Dou X, Zhao Y, Wang X, Zhao C, Xia J, Xing C, He S, Feng C. Chiral Supramolecular Nanofibers Regulated Tumor-Derived Exosomes Secretion for Constructing an Anti-Tumor Extracellular Microenvironment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308335. [PMID: 38420895 DOI: 10.1002/smll.202308335] [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: 09/21/2023] [Revised: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Tumor-derived exosomes (TDEs) induced extracellular microenvironment has recently been validated to be critical for tumor progression and metastasis, however, remodeling it for oncotherapy still remains a major challenge due to difficulty in regulation of TDEs secretion. Herein, the supramolecular chiral nanofibers, composed of L/D-phenylalanine derivates (L/D-Phe) and linear hyaluronic acid (HA), are successfully employed to construct TDEs induced anti-tumor extracellular microenvironment. The left-handed L-Phe @HA nanofibers significantly inhibit TDEs secretion into extracellular microenvironment, which results in suppression of tumor proliferation and metastasis in vitro and vivo. Biological assays and theoretical modeling reveal that these results are mainly attributed to strong adsorption of the key exosomes transporters (Ras-related protein Rab-27A and synaptosome-associated protein 23) on left-handed L-Phe @HA nanofibers via enhanced stereoselective interaction, leading to degradation and phosphorylated dropping of exosomes transporters. Subsequently, transfer function of exosomes transporters is limited, which causes remarkable inhibition of TDEs secretion. These findings provide a promising novel insight of chiral functional materials to establish an anti-tumor extracellular microenvironment via regulation of TDEs secretion.
Collapse
Affiliation(s)
- Beibei Wu
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Xiaoqiu Dou
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Yu Zhao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Xueqian Wang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Changli Zhao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Jingyi Xia
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Chao Xing
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Sijia He
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| | - Chuanliang Feng
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200230, P. R. China
| |
Collapse
|
29
|
Wang Y. The interplay of exercise and polyphenols in cancer treatment: A focus on oxidative stress and antioxidant mechanisms. Phytother Res 2024; 38:3459-3488. [PMID: 38690720 DOI: 10.1002/ptr.8215] [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: 02/01/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 05/02/2024]
Abstract
Exercise has been demonstrated to induce an elevated production of free radicals, leading to the onset of oxidative stress. Numerous studies highlight the positive impacts of aerobic exercise, primarily attributed to the increase in overall antioxidant capacity. The evidence suggests that engaging in aerobic exercise contributes to a reduction in the likelihood of advanced cancer and mortality. Oxidative stress occurs when there is an imbalance between the generation of free radicals and the collective antioxidant defense system, encompassing both enzymatic and nonenzymatic antioxidants. Typically, oxidative stress triggers the formation of reactive oxygen or nitrogen species, instigating or advancing various issues in cancers and other diseases. The pro-oxidant-antioxidant balance serves as a direct measure of this imbalance in oxidative stress. Polyphenols contain a variety of bioactive compounds, including flavonoids, flavanols, and phenolic acids, conferring antioxidant properties. Previous research highlights the potential of polyphenols as antioxidants, with documented effects on reducing cancer risk by influencing processes such as proliferation, angiogenesis, and metastasis. This is primarily attributed to their recognized antioxidant capabilities. Considering the extensive array of signaling pathways associated with exercise and polyphenols, this overview will specifically focus on oxidative stress, the antioxidant efficacy of polyphenols and exercise, and their intricate interplay in cancer treatment.
Collapse
Affiliation(s)
- Yubing Wang
- College of Physical Education, Qilu Normal University, Jinan, Shandong, China
| |
Collapse
|
30
|
Yue Y, She X, Ding W, Chen S, Xiao Q, Pan B, Zhou L, Yin Y, Li Y, Wang S, Xu M. A novel Senescence-Based prognostic model unveils tumor interactions and drug resistance in colorectal cancer. Int Immunopharmacol 2024; 134:112197. [PMID: 38733826 DOI: 10.1016/j.intimp.2024.112197] [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: 02/17/2024] [Revised: 04/12/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND In China, CRC incidence is escalating. The main hurdles are heterogeneity and drug resistance. This research delves into cellular senescence in CRC, aiming to devise a prognostic model and pinpoint mechanisms impacting drug resistance. METHODS Mendelian randomization (MR) analysis confirmed the association between CRC and cellular aging. The Cancer Genome Atlas (TCGA)-CRC data served as the training set, with GSE38832 and GSE39582 as validation sets. Various bioinformatics methods were employed to construct and validate a risk model. CRC cells with NADPH Oxidase 4 (NOX4) knockout were generated using CRISPR-Cas9 technology. Protein blotting and colony formation assays elucidated the role of NOX4 in CRC cell aging and drug resistance. RESULTS A prognostic model, derived from dataset analysis, uncovered a link between high-risk groups and cancer progression. Notable differences in the tumor microenvironment were observed between risk groups. Finally, NOX4 was found to be linked with aging and drug resistance in CRC. CONCLUSION This research presents a novel senescence-based CRC prognosis model. It identifies NOX4's role in CRC drug resistance, suggesting it is a potential treatment target.
Collapse
Affiliation(s)
- Yanzhe Yue
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Xiangjian She
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Wenbo Ding
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Shuyu Chen
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Qianni Xiao
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Bei Pan
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Linpeng Zhou
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China
| | - Yujuan Yin
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Youyue Li
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China
| | - Shukui Wang
- General Clinical Research Center, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, Jiangsu, China; School of Basic-Medicine and Clinical Pharmacy, Nanjing First Hospital, China Pharmaceutical University, 211198, Nanjing, Jiangsu, China; Jiangsu Collaborative Innovation Center on Cancer Personalized Medicine, Nanjing Medical University, 211166, Nanjing, Jiangsu, China.
| | - Mu Xu
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, 211166, Nanjing, China.
| |
Collapse
|
31
|
Yao L, Zhu X, Shan Y, Zhang L, Yao J, Xiong H. Recent Progress in Anti-Tumor Nanodrugs Based on Tumor Microenvironment Redox Regulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310018. [PMID: 38269480 DOI: 10.1002/smll.202310018] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/30/2023] [Indexed: 01/26/2024]
Abstract
The growth state of tumor cells is strictly affected by the specific abnormal redox status of the tumor microenvironment (TME). Moreover, redox reactions at the biological level are also central and fundamental to essential energy metabolism reactions in tumors. Accordingly, anti-tumor nanodrugs targeting the disruption of this abnormal redox homeostasis have become one of the hot spots in the field of nanodrugs research due to the effectiveness of TME modulation and anti-tumor efficiency mediated by redox interference. This review discusses the latest research results of nanodrugs in anti-tumor therapy, which regulate the levels of oxidants or reductants in TME through a variety of therapeutic strategies, ultimately breaking the original "stable" redox state of the TME and promoting tumor cell death. With the gradual deepening of study on the redox state of TME and the vigorous development of nanomaterials, it is expected that more anti-tumor nano drugs based on tumor redox microenvironment regulation will be designed and even applied clinically.
Collapse
Affiliation(s)
- Lan Yao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Xiang Zhu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Yunyi Shan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Liang Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Jing Yao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| | - Hui Xiong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, Department of Pharmaceutics, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, P. R. China
| |
Collapse
|
32
|
Streich S, Higuchi J, Opalińska A, Wojnarowicz J, Giovanoli P, Łojkowski W, Buschmann J. Ultrasonic Coating of Poly(D,L-lactic acid)/Poly(lactic-co-glycolic acid) Electrospun Fibers with ZnO Nanoparticles to Increase Angiogenesis in the CAM Assay. Biomedicines 2024; 12:1155. [PMID: 38927362 PMCID: PMC11201106 DOI: 10.3390/biomedicines12061155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/07/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Critical-size bone defects necessitate bone void fillers that should be integrated well and be easily vascularized. One viable option is to use a biocompatible synthetic polymer and sonocoat it with zinc oxide (ZnO) nanoparticles (NPs). However, the ideal NP concentration and size must be assessed because a high dose of ZnO NPs may be toxic. Electrospun PDLLA/PLGA scaffolds were produced with different concentrations (0.5 or 1.0 s of sonocoating) and sizes of ZnO NPs (25 nm and 70 nm). They were characterized by SEM, EDX, ICP-OES, and the water contact angle. Vascularization and integration into the surrounding tissue were assessed with the CAM assay in the living chicken embryo. SEM, EDX, and ICP-OES confirmed the presence of ZnO NPs on polymer fibers. Sonocoated ZnO NPs lowered the WCA compared with the control. Smaller NPs were more pro-angiogenic exhibiting a higher vessel density than the larger NPs. At a lower concentration, less but larger vessels were visible in an environment with a lower cell density. Hence, the favored combination of smaller ZnO NPs at a lower concentration sonocoated on PDLLA/PLGA electrospun meshes leads to an advanced state of tissue integration and vascularization, providing a valuable synthetic bone graft to be used in clinics in the future.
Collapse
Affiliation(s)
- Selina Streich
- Medical Faculty, University of Zurich, Campus Irchel, 8006 Zurich, Switzerland;
- Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Julia Higuchi
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Sokolowska 29/37, 01-142 Warsaw, Poland; (J.H.); (A.O.); (J.W.); (W.Ł.)
| | - Agnieszka Opalińska
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Sokolowska 29/37, 01-142 Warsaw, Poland; (J.H.); (A.O.); (J.W.); (W.Ł.)
| | - Jacek Wojnarowicz
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Sokolowska 29/37, 01-142 Warsaw, Poland; (J.H.); (A.O.); (J.W.); (W.Ł.)
| | - Pietro Giovanoli
- Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Witold Łojkowski
- Laboratory of Nanostructures, Institute of High Pressure Physics, Polish Academy of Sciences, Sokolowska 29/37, 01-142 Warsaw, Poland; (J.H.); (A.O.); (J.W.); (W.Ł.)
| | - Johanna Buschmann
- Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland;
| |
Collapse
|
33
|
De Leo A, Ugolini A, Yu X, Scirocchi F, Scocozza D, Peixoto B, Pace A, D'Angelo L, Liu JKC, Etame AB, Rughetti A, Nuti M, Santoro A, Vogelbaum MA, Conejo-Garcia JR, Rodriguez PC, Veglia F. Glucose-driven histone lactylation promotes the immunosuppressive activity of monocyte-derived macrophages in glioblastoma. Immunity 2024; 57:1105-1123.e8. [PMID: 38703775 PMCID: PMC11114377 DOI: 10.1016/j.immuni.2024.04.006] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 12/08/2023] [Accepted: 04/09/2024] [Indexed: 05/06/2024]
Abstract
Immunosuppressive macrophages restrict anti-cancer immunity in glioblastoma (GBM). Here, we studied the contribution of microglia (MGs) and monocyte-derived macrophages (MDMs) to immunosuppression and mechanisms underlying their regulatory function. MDMs outnumbered MGs at late tumor stages and suppressed T cell activity. Molecular and functional analysis identified a population of glycolytic MDM expressing GLUT1 with potent immunosuppressive activity. GBM-derived factors promoted high glycolysis, lactate, and interleukin-10 (IL-10) production in MDMs. Inhibition of glycolysis or lactate production in MDMs impaired IL-10 expression and T cell suppression. Mechanistically, intracellular lactate-driven histone lactylation promoted IL-10 expression, which was required to suppress T cell activity. GLUT1 expression on MDMs was induced downstream of tumor-derived factors that activated the PERK-ATF4 axis. PERK deletion in MDM abrogated histone lactylation, led to the accumulation of intratumoral T cells and tumor growth delay, and, in combination with immunotherapy, blocked GBM progression. Thus, PERK-driven glucose metabolism promotes MDM immunosuppressive activity via histone lactylation.
Collapse
Affiliation(s)
- Alessandra De Leo
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Alessio Ugolini
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA; Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Xiaoqing Yu
- Department of Biostatistics and Bioinformatic, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Fabio Scirocchi
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Delia Scocozza
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Barbara Peixoto
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Angelica Pace
- Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Luca D'Angelo
- Department of Human Neurosciences, Neurosurgery Division, "Sapienza" University, AOU Policlinico Umberto I, Rome, Italy
| | - James K C Liu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Arnold B Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Aurelia Rughetti
- Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Marianna Nuti
- Department of Experimental Medicine "Sapienza" University of Rome, Rome, Italy
| | - Antonio Santoro
- Department of Human Neurosciences, Neurosurgery Division, "Sapienza" University, AOU Policlinico Umberto I, Rome, Italy
| | | | - Jose R Conejo-Garcia
- Department of Integrative immunobiology, Duke School of Medicine, Durham, NC, USA
| | - Paulo C Rodriguez
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Filippo Veglia
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Department of Neuro-Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Develin A, Fuglestad B. Inositol Hexaphosphate as an Inhibitor and Potential Regulator of p47 phox Membrane Anchoring. Biochemistry 2024; 63:1097-1106. [PMID: 38669178 PMCID: PMC11080064 DOI: 10.1021/acs.biochem.4c00117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/31/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024]
Abstract
As a key component for NADPH oxidase 2 (NOX2) activation, the peripheral membrane protein p47phox translocates a cytosolic activating complex to the membrane through its PX domain. This study elucidates a potential regulatory mechanism of p47phox recruitment and NOX2 activation by inositol hexaphosphate (IP6). Through NMR, fluorescence polarization, and FRET experimental results, IP6 is shown to be capable of breaking the lipid binding and membrane anchoring events of p47phox-PX with low micromolar potency. Other phosphorylated inositol species such as IP5(1,3,4,5,6), IP4(1,3,4,5), and IP3(1,3,4) show weaker binding and no ability to inhibit lipid interactions in physiological concentration ranges. The low micromolar potency of IP6 inhibition of the p47phox membrane anchoring suggests that physiologically relevant concentrations of IP6 serve as regulators, as seen in other membrane anchoring domains. The PX domain of p47phox is known to be promiscuous to a variety of phosphatidylinositol phosphate (PIP) lipids, and this regulation may help target the domain only to the membranes most highly enriched with the highest affinity PIPs, such as the phagosomal membrane, while preventing aberrant binding to other membranes with high and heterogeneous PIP content, such as the plasma membrane. This study provides insight into a potential novel regulatory mechanism behind NOX2 activation and reveals a role for small-molecule regulation in this important NOX2 activator.
Collapse
Affiliation(s)
- Angela
M. Develin
- Department
of Chemistry, Virginia Commonwealth University, Richmond, Virginia 22384, United States
| | - Brian Fuglestad
- Department
of Chemistry, Virginia Commonwealth University, Richmond, Virginia 22384, United States
- Institute
for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, Virginia 23219, United States
| |
Collapse
|
35
|
Dakroub A, Dbouk A, Asfour A, Nasser SA, El-Yazbi AF, Sahebkar A, Eid AA, Iratni R, Eid AH. C-peptide in diabetes: A player in a dual hormone disorder? J Cell Physiol 2024; 239:e31212. [PMID: 38308646 DOI: 10.1002/jcp.31212] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/05/2024]
Abstract
C-peptide, a byproduct of insulin synthesis believed to be biologically inert, is emerging as a multifunctional molecule. C-peptide serves an anti-inflammatory and anti-atherogenic role in type 1 diabetes mellitus (T1DM) and early T2DM. C-peptide protects endothelial cells by activating AMP-activated protein kinase α, thus suppressing the activity of NAD(P)H oxidase activity and reducing reactive oxygen species (ROS) generation. It also prevents apoptosis by regulating hyperglycemia-induced p53 upregulation and mitochondrial adaptor p66shc overactivation, as well as reducing caspase-3 activity and promoting expression of B-cell lymphoma-2. Additionally, C-peptide suppresses platelet-derived growth factor (PDGF)-beta receptor and p44/p42 mitogen-activated protein (MAP) kinase phosphorylation to inhibit vascular smooth muscle cells (VSMC) proliferation. It also diminishes leukocyte adhesion by virtue of its capacity to abolish nuclear factor kappa B (NF-kB) signaling, a major pro-inflammatory cascade. Consequently, it is envisaged that supplementation of C-peptide in T1DM might ameliorate or even prevent end-organ damage. In marked contrast, C-peptide increases monocyte recruitment and migration through phosphoinositide 3-kinase (PI-3 kinase)-mediated pathways, induces lipid accumulation via peroxisome proliferator-activated receptor γ upregulation, and stimulates VSMC proliferation and CD4+ lymphocyte migration through Src-kinase and PI-3K dependent pathways. Thus, it promotes atherosclerosis and microvascular damage in late T2DM. Indeed, C-peptide is now contemplated as a potential biomarker for insulin resistance in T2DM and linked to increased coronary artery disease risk. This shift in the understanding of the pathophysiology of diabetes from being a single hormone deficiency to a dual hormone disorder warrants a careful consideration of the role of C-peptide as a unique molecule with promising diagnostic, prognostic, and therapeutic applications.
Collapse
Affiliation(s)
- Ali Dakroub
- St. Francis Hospital and Heart Center, Roslyn, New York, USA
| | - Ali Dbouk
- Department of Medicine, Saint-Joseph University Medical School, Hotel-Dieu de France Hospital, Beirut, Lebanon
| | - Aref Asfour
- Leeds Teaching Hospitals NHS Trust, West Yorkshire, United Kingdom
| | | | - Ahmed F El-Yazbi
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Egypt
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, UAE
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
36
|
Han D, Ding B, Zheng P, Yuan M, Bian Y, Chen H, Wang M, Chang M, Kheraif AAA, Ma P, Lin J. NADPH Oxidase-Like Nanozyme for High-Efficiency Tumor Therapy Through Increasing Glutathione Consumption and Blocking Glutathione Regeneration. Adv Healthc Mater 2024; 13:e2303309. [PMID: 38214472 DOI: 10.1002/adhm.202303309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/18/2023] [Indexed: 01/13/2024]
Abstract
To counteract the high level of reactive oxygen species (ROS) caused by rapid growth, tumor cells resist oxidative stress by accelerating the production and regeneration of intracellular glutathione (GSH). Numerous studies focus on the consumption of GSH, but the regeneration of GSH will enhance the reduction level of tumor cells to resist oxidative stress. Therefore, inhibiting the regeneration of GSH; while, consuming GSH is of great significance for breaking the redox balance of tumor cells. Herein, a simple termed MnOx-coated Au (AMO) nanoflower, as a nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) nanoenzyme, is reported for efficient tumor therapy. Au nanoparticles exhibit the capability to catalyze the oxidation of NADPH, hindering GSH regeneration; while, concurrently functioning as a photothermal agent. During the process of eliminating intracellular GSH, MnOx releases Mn2+ that subsequently engages in Fenton-like reactions, ultimately facilitating the implementation of chemodynamic therapy (CDT). Overall, this NOX enzyme-based nanoplatform enhances ROS generation and disrupts the state of reduction equilibrium, inducing apoptosis and ferroptosis by blocking GSH regeneration and increasing GSH consumption, thereby achieving collaborative treatments involving photothermal therapy (PTT), CDT, and catalytic therapy. This research contributes to NADPH and GSH targeted tumor therapy and showcases the potential of nanozymes.
Collapse
Affiliation(s)
- Di Han
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Binbin Ding
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Pan Zheng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- Key Laboratory of Superlight Materials and Surface Technology of Ministry of Education, College of Material Sciences and Chemical Engineering, Harbin Engineering University, Harbin, 150001, P. R. China
| | - Meng Yuan
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Yulong Bian
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Hao Chen
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Meifang Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Mengyu Chang
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, 77030, USA
| | - Abdulaziz A Al Kheraif
- Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh, 12372, Saudi Arabia
| | - Ping'an Ma
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
37
|
Liao P, Chen L, Zhou H, Mei J, Chen Z, Wang B, Feng JQ, Li G, Tong S, Zhou J, Zhu S, Qian Y, Zong Y, Zou W, Li H, Zhang W, Yao M, Ma Y, Ding P, Pang Y, Gao C, Mei J, Zhang S, Zhang C, Liu D, Zheng M, Gao J. Osteocyte mitochondria regulate angiogenesis of transcortical vessels. Nat Commun 2024; 15:2529. [PMID: 38514612 PMCID: PMC10957947 DOI: 10.1038/s41467-024-46095-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 02/13/2024] [Indexed: 03/23/2024] Open
Abstract
Transcortical vessels (TCVs) provide effective communication between bone marrow vascular system and external circulation. Although osteocytes are in close contact with them, it is not clear whether osteocytes regulate the homeostasis of TCVs. Here, we show that osteocytes maintain the normal network of TCVs by transferring mitochondria to the endothelial cells of TCV. Partial ablation of osteocytes causes TCV regression. Inhibition of mitochondrial transfer by conditional knockout of Rhot1 in osteocytes also leads to regression of the TCV network. By contrast, acquisition of osteocyte mitochondria by endothelial cells efficiently restores endothelial dysfunction. Administration of osteocyte mitochondria resultes in acceleration of the angiogenesis and healing of the cortical bone defect. Our results provide new insights into osteocyte-TCV interactions and inspire the potential application of mitochondrial therapy for bone-related diseases.
Collapse
Affiliation(s)
- Peng Liao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Long Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hao Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jiong Mei
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziming Chen
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Bingqi Wang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jerry Q Feng
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, China
| | - Guangyi Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Sihan Tong
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Zhou
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyuan Zhu
- Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Qian
- Department of Orthopedics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia
| | - Weiguo Zou
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenkan Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Meng Yao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiyang Ma
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Ding
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yidan Pang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuan Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialun Mei
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Senyao Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Delin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Minghao Zheng
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Western Australia, Australia.
- Perron Institute for Neurological and Translational Science, Nedlands, Western Australia, Australia.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
38
|
Rasouli M, Fattahi R, Nuoroozi G, Zarei-Behjani Z, Yaghoobi M, Hajmohammadi Z, Hosseinzadeh S. The role of oxygen tension in cell fate and regenerative medicine: implications of hypoxia/hyperoxia and free radicals. Cell Tissue Bank 2024; 25:195-215. [PMID: 37365484 DOI: 10.1007/s10561-023-10099-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
Oxygen pressure plays an integral role in regulating various aspects of cellular biology. Cell metabolism, proliferation, morphology, senescence, metastasis, and angiogenesis are some instances that are affected by different tensions of oxygen. Hyperoxia or high oxygen concentration, enforces the production of reactive oxygen species (ROS) that disturbs physiological homeostasis, and consequently, in the absence of antioxidants, cells and tissues are directed to an undesired fate. On the other side, hypoxia or low oxygen concentration, impacts cell metabolism and fate strongly through inducing changes in the expression level of specific genes. Thus, understanding the precise mechanism and the extent of the implication of oxygen tension and ROS in biological events is crucial to maintaining the desired cell and tissue function for application in regenerative medicine strategies. Herein, a comprehensive literature review has been performed to find out the impacts of oxygen tensions on the various behaviors of cells or tissues.
Collapse
Affiliation(s)
- Mehdi Rasouli
- Student Research Committee, Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Fattahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Ghader Nuoroozi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Zeinab Zarei-Behjani
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maliheh Yaghoobi
- Engineering Department, Faculty of Chemical Engineering, Zanjan University, Zanjan, Iran
| | - Zeinab Hajmohammadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran.
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Neal CJ, Kolanthai E, Wei F, Coathup M, Seal S. Surface Chemistry of Biologically Active Reducible Oxide Nanozymes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2211261. [PMID: 37000888 DOI: 10.1002/adma.202211261] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/15/2023] [Indexed: 06/19/2023]
Abstract
Reducible metal oxide nanozymes (rNZs) are a subject of intense recent interest due to their catalytic nature, ease of synthesis, and complex surface character. Such materials contain surface sites which facilitate enzyme-mimetic reactions via substrate coordination and redox cycling. Further, these surface reactive sites are shown to be highly sensitive to stresses within the nanomaterial lattice, the physicochemical environment, and to processing conditions occurring as part of their syntheses. When administered in vivo, a complex protein corona binds to the surface, redefining its biological identity and subsequent interactions within the biological system. Catalytic activities of rNZs each deliver a differing impact on protein corona formation, its composition, and in turn, their recognition, and internalization by host cells. Improving the understanding of the precise principles that dominate rNZ surface-biomolecule adsorption raises the question of whether designer rNZs can be engineered to prevent corona formation, or indeed to produce "custom" protein coronas applied either in vitro, and preadministration, or formed immediately upon their exposure to body fluids. Here, fundamental surface chemistry processes and their implications in rNZ material performance are considered. In particular, material structures which inform component adsorption from the application environment, including substrates for enzyme-mimetic reactions are discussed.
Collapse
Affiliation(s)
- Craig J Neal
- Advanced Materials Processing and Analysis Center, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Elayaraja Kolanthai
- Advanced Materials Processing and Analysis Center, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Fei Wei
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Melanie Coathup
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| | - Sudipta Seal
- Advanced Materials Processing and Analysis Center, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
- Biionix Cluster, Department of Internal Medicine, College of Medicine, University of Central Florida, Orlando, FL, 32827, USA
| |
Collapse
|
40
|
Mamun MAA, Rakib A, Mandal M, Kumar S, Singla B, Singh UP. Polyphenols: Role in Modulating Immune Function and Obesity. Biomolecules 2024; 14:221. [PMID: 38397458 PMCID: PMC10887194 DOI: 10.3390/biom14020221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/02/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Polyphenols, long-used components of medicinal plants, have drawn great interest in recent years as potential therapeutic agents because of their safety, efficacy, and wide range of biological effects. Approximately 75% of the world's population still use plant-based medicinal compounds, indicating the ongoing significance of phytochemicals for human health. This study emphasizes the growing body of research investigating the anti-adipogenic and anti-obesity functions of polyphenols. The functions of polyphenols, including phenylpropanoids, flavonoids, terpenoids, alkaloids, glycosides, and phenolic acids, are distinct due to changes in chemical diversity and structural characteristics. This review methodically investigates the mechanisms by which naturally occurring polyphenols mediate obesity and metabolic function in immunomodulation. To this end, hormonal control of hunger has the potential to inhibit pro-obesity enzymes such as pancreatic lipase, the promotion of energy expenditure, and the modulation of adipocytokine production. Specifically, polyphenols affect insulin, a hormone that is essential for regulating blood sugar, and they also play a role, in part, in a complex web of factors that affect the progression of obesity. This review also explores the immunomodulatory properties of polyphenols, providing insight into their ability to improve immune function and the effects of polyphenols on gut health, improving the number of commensal bacteria, cytokine production suppression, and immune cell mediation, including natural killer cells and macrophages. Taken together, continuous studies are required to understand the prudent and precise mechanisms underlying polyphenols' therapeutic potential in obesity and immunomodulation. In the interim, this review emphasizes a holistic approach to health and promotes the consumption of a wide range of foods and drinks high in polyphenols. This review lays the groundwork for future developments, indicating that the components of polyphenols and their derivatives may provide the answer to urgent worldwide health issues. This compilation of the body of knowledge paves the way for future discoveries in the global treatment of pressing health concerns in obesity and metabolic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Udai P. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN 38163, USA; (M.A.A.M.); (A.R.); (M.M.); (S.K.); (B.S.)
| |
Collapse
|
41
|
Balasubramanian P, Vijayarangam V, Deviparasakthi MKG, Palaniyandi T, Ravi M, Natarajan S, Viswanathan S, Baskar G, Wahab MRA, Surendran H. Implications and progression of peroxiredoxin 2 (PRDX2) in various human diseases. Pathol Res Pract 2024; 254:155080. [PMID: 38219498 DOI: 10.1016/j.prp.2023.155080] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/24/2023] [Accepted: 12/30/2023] [Indexed: 01/16/2024]
Abstract
Peroxiredoxin 2 (PRDX2), a characteristic 2-Cys enzyme is one of the foremost effective scavenger proteins against reactive oxygen species (ROS) and hydrogen peroxide (H2O2) defending cells against oxidative stress. Dysregulation of this antioxidant raises the quantity of ROS and oxidative stress implicated in several diseases. PRDX2 lowers the generation of ROS that takes part in controlling several signalling pathways occurring in neurons, protecting them from stress caused by oxidation and an inflammatory harm. Depending on the aetiological variables, the kind of cancer, and the stage of tumour development, PRDX2 may behave either as an onco-suppressor or a promoter. However, overexpression of PRDX2 may be linked to the development of numerous cancers, including those of the colon, cervix, breast, and prostate. PRDX2 also plays a beneficial effect in inflammatory diseases. PRDX2 being a thiol-specific peroxidase, is known to control proinflammatory reactions. The spilling of PRDX2, on the other hand, accelerates cognitive impairment following a stroke by triggering an inflammatory reflex. PRDX2 expression patterns in vascular cells tend to be crucial to its involvement in cardiovascular diseases. In vascular smooth muscle cells, if the protein tyrosine phosphatase is restricted, PRDX2 could avoid the neointimal thickening which relies on platelet derived growth factor (PDGF), a vital component of vascular remodelling. A proper PRDX2 balance is therefore crucial. The imbalance causes a number of illnesses, including cancers, inflammatory diseases, cardiovascular ailments, and neurological and neurodegenerative problems which are discussed in this review.
Collapse
Affiliation(s)
| | - Varshini Vijayarangam
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | | | - Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India; Department of Anatomy, Biomedical Research Unit and Laboratory Animal Centre, Saveetha Dental College and Hospital, SIMATS, Saveetha University, Chennai, India.
| | - Maddaly Ravi
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Sudhakar Natarajan
- Department of Tuberculosis, ICMR - National Institute for Research in Tuberculosis (NIRT), Chennai, India
| | - Sandhiya Viswanathan
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | | | - Hemapreethi Surendran
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Chennai, India
| |
Collapse
|
42
|
Shah R, Ibis B, Kashyap M, Boussiotis VA. The role of ROS in tumor infiltrating immune cells and cancer immunotherapy. Metabolism 2024; 151:155747. [PMID: 38042522 PMCID: PMC10872310 DOI: 10.1016/j.metabol.2023.155747] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/16/2023] [Accepted: 11/29/2023] [Indexed: 12/04/2023]
Abstract
Reactive oxygen species (ROS) are a group of short-lived highly reactive molecules formed intracellularly from molecular oxygen. ROS can alter biochemical, transcriptional, and epigenetic programs and have an indispensable role in cellular function. In immune cells, ROS are mediators of specialized functions such as phagocytosis, antigen presentation, activation, cytolysis, and differentiation. ROS have a fundamental role in the tumor microenvironment (TME) where they are produced by immune cell-intrinsic and -extrinsic mechanisms. ROS can act as a double-edged sword with short exposures leading to activation in various innate and adaptative immune cells, and prolonged exposures, unopposed by redox balancing antioxidants leading to exhaustion, immunosuppression, and unresponsiveness to cancer immunotherapy. Due to its plasticity and impact on the anti-tumor function of immune cells, attempts are currently in process to harness ROS biology with the purpose to improve contemporary strategies of cancer immunotherapy. Here, we provide a short overview how ROS and various antioxidant systems impact on the function of innate and adaptive immune system cells with emphasis on the TME and immune-based therapies for cancer.
Collapse
Affiliation(s)
- Rushil Shah
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Betul Ibis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Monisha Kashyap
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America; Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States of America.
| |
Collapse
|
43
|
Kumari R, Syeda S, Shrivastava A. Nature's Elixir for Cancer Treatment: Targeting Tumor-induced Neovascularization. Curr Med Chem 2024; 31:5281-5304. [PMID: 38425113 DOI: 10.2174/0109298673282525240222050051] [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: 10/17/2023] [Revised: 01/20/2024] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
Angiogenesis, a multistep process, involves sprouting of new vessels from the pre-existing vessels in response to a stimulus in its microenvironment. Normally, angiogenesis is important for tissue maintenance and homeostasis, however it is also known to be associated with various pathologies, including cancer. Importantly, neovascularization is very crucial for tumors to grow and metastasize since it allows delivery of oxygen and nutrients as well as promotes tumor cell dissemination to distant sites. Activation of angiogenic switch is a consequence of imbalance in pro- as well as anti-angiogenic factors, that are immensely impacted by reactive oxygen species and epigenetic regulation. Several reports have suggested that angiogenic inhibitors significantly inhibit tumor growth. Therefore, anti-angiogenic therapy has gained substantial attention and has been considered a rational approach in cancer therapeutics. In this line, several anti- angiogenic drugs have been approved, however, their long term usage caused several side effects. In view of this, researchers switched to plant-based natural compounds for identifying safe and cost-effective anti-angiogenic drugs. Of note, various phytochemicals have been evaluated to reduce tumor growth by inhibiting tumor-induced angiogenesis. Moreover, the implication of nano-carriers to enhance the bioavailability of phytochemicals has proven to be more efficient anti-cancer agents. The present review highlights the existing knowledge on tumor-induced neovascularization and its regulation at the epigenetic level. Further, we emphasize the inhibitory effect of phytochemicals on tumor- induced angiogenesis that will open up new avenues in cancer therapeutics.
Collapse
Affiliation(s)
- Rani Kumari
- Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Saima Syeda
- Department of Zoology, University of Delhi, Delhi, 110007, India
| | - Anju Shrivastava
- Department of Zoology, University of Delhi, Delhi, 110007, India
| |
Collapse
|
44
|
Hajibabaie F, Abedpoor N, Haghjooy Javanmard S, Hasan A, Sharifi M, Rahimmanesh I, Shariati L, Makvandi P. The molecular perspective on the melanoma and genome engineering of T-cells in targeting therapy. ENVIRONMENTAL RESEARCH 2023; 237:116980. [PMID: 37648188 DOI: 10.1016/j.envres.2023.116980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/19/2023] [Accepted: 08/23/2023] [Indexed: 09/01/2023]
Abstract
Melanoma, an aggressive malignant tumor originating from melanocytes in humans, is on the rise globally, with limited non-surgical treatment options available. Recent advances in understanding the molecular and cellular mechanisms underlying immune escape, tumorigenesis, drug resistance, and cancer metastasis have paved the way for innovative therapeutic strategies. Combination therapy targeting multiple pathways simultaneously has been shown to be promising in treating melanoma, eliciting favorable responses in most melanoma patients. CAR T-cells, engineered to overcome the limitations of human leukocyte antigen (HLA)-dependent tumor cell detection associated with T-cell receptors, offer an alternative approach. By genetically modifying apheresis-collected allogeneic or autologous T-cells to express chimeric antigen receptors, CAR T-cells can appreciate antigens on cell surfaces independently of major histocompatibility complex (MHC), providing a significant cancer cell detection advantage. However, identifying the most effective target antigen is the initial step, as it helps mitigate the risk of toxicity due to "on-target, off-tumor" and establishes a targeted therapeutic strategy. Furthermore, evaluating signaling pathways and critical molecules involved in melanoma pathogenesis remains insufficient. This study emphasizes the novel approaches of CAR T-cell immunoediting and presents new insights into the molecular signaling pathways associated with melanoma.
Collapse
Affiliation(s)
- Fatemeh Hajibabaie
- Department of Biology, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran; Department of Medical Biotechnology, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Navid Abedpoor
- Department of Sports Physiology, Faculty of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran; Department of Medical Biotechnology, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, 2713, Qatar; Biomedical Research Center, Qatar University, Doha, 2713, Qatar.
| | - Mehran Sharifi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran; Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| |
Collapse
|
45
|
Song D, Yang X, Chen Y, Hu P, Zhang Y, Zhang Y, Liang N, Xie J, Qiao L, Deng G, Chen F, Zhang J. Advances in anti-tumor based on various anaerobic bacteria and their derivatives as drug vehicles. Front Bioeng Biotechnol 2023; 11:1286502. [PMID: 37854883 PMCID: PMC10579911 DOI: 10.3389/fbioe.2023.1286502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023] Open
Abstract
Cancer therapies, such as chemotherapy and radiotherapy, are often unsatisfactory due to several limitations, including drug resistance, inability to cross biological barriers, and toxic side effects on the body. These drawbacks underscore the need for alternative treatments that can overcome these challenges and provide more effective and safer options for cancer patients. In recent years, the use of live bacteria, engineered bacteria, or bacterial derivatives to deliver antitumor drugs to specific tumor sites for controlled release has emerged as a promising therapeutic tool. This approach offers several advantages over traditional cancer therapies, including targeted drug delivery and reduced toxicity to healthy tissues. Ongoing research in this field holds great potential for further developing more efficient and personalized cancer therapies, such as E. coli, Salmonella, Listeria, and bacterial derivatives like outer membrane vesicles (OMVs), which can serve as vehicles for drugs, therapeutic proteins, or antigens. In this review, we describe the advances, challenges, and future directions of research on using live bacteria or OMVs as carriers or components derived from bacteria of delivery systems for cancer therapy.
Collapse
Affiliation(s)
- Daichen Song
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaofan Yang
- School of Clinical Medicine, Jining Medical University, Jining, China
| | - Yanfei Chen
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Pingping Hu
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yingying Zhang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yan Zhang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Ning Liang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jian Xie
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Lili Qiao
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Guodong Deng
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Fangjie Chen
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiandong Zhang
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Department of Oncology, Shandong Lung Cancer Institute, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
46
|
Palanissami G, Paul SF. AGEs and RAGE: metabolic and molecular signatures of the glycation-inflammation axis in malignant or metastatic cancers. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:812-849. [PMID: 37970208 PMCID: PMC10645465 DOI: 10.37349/etat.2023.00170] [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: 03/28/2023] [Accepted: 06/12/2023] [Indexed: 11/17/2023] Open
Abstract
From attributing mutations to cancers with the advent of cutting-edge genetic technology in recent decades, to re-searching the age-old theory of intrinsic metabolic shift of cancers (Warburg's glycolysis), the quest for a precise panacea for mainly the metastatic cancers, remains incessant. This review delineates the advanced glycation end product (AGE)-receptor for AGE (RAGE) pathway driven intricate oncogenic cues, budding from the metabolic (glycolytic) reliance of tumour cells, branching into metastatic emergence of malignancies. Strong AGE-RAGE concomitance in metastasis, chemo-resistance and cancer resurgence adversely incite disease progression and patient mortality. At the conjunction of metabolic and metastatic shift of cancers, are the "glycolytically" generated AGEs and AGE-activated RAGE, instigating aberrant molecular pathways, culminating in aggressive malignancies. AGEs as by-products of metabolic insurgence, modify the metabolome, epigenome and microbiome, besides coercing the inter-, intra- and extra-cellular micro-milieu conducive for oncogenic events like epithelial-mesenchymal transition (EMT). AGE-RAGE synergistically elicit ATP surge for surplus energy, autophagy for apoptotic evasion and chemo-resistance, insulin-like growth factor 1 (IGF-1) for meta-inflammation and angiogenesis, high mobility group box-1 (HMGB1) for immune tolerance, S100 proteins for metastasis, and p53 protein attenuation for tumour suppression. AGEs are pronouncedly reported in invasive forms of breast, prostate, colon and pancreatic cancers, higher in patients with cancer than healthy counterparts, and higher in advanced stage than localized phase. Hence, the investigation of person-specific presence of AGEs, soluble RAGE and AGE-activated RAGE can be advocated as impending bio-markers for diagnostic, prognostic and therapeutic purposes, to predict cancer risk in patients with diabetes, obesity, metabolic syndrome as well as general population, to monitor prognosis and metastasis in patients with cancer, and to reckon complications in cancer survivors. Furthermore, clinical reports of exogenous (dietary) and endogenous (internally formed) AGEs in cancer patients, and contemporary clinical trials involving AGE-RAGE axis in cancer are underlined with theranostic implications.
Collapse
Affiliation(s)
- Gowri Palanissami
- Department of Human Genetics, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Porur, Chennai 600 116, Tamil Nadu, India
| | - Solomon F.D. Paul
- Department of Human Genetics, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Porur, Chennai 600 116, Tamil Nadu, India
| |
Collapse
|
47
|
Cipriano A, Viviano M, Feoli A, Milite C, Sarno G, Castellano S, Sbardella G. NADPH Oxidases: From Molecular Mechanisms to Current Inhibitors. J Med Chem 2023; 66:11632-11655. [PMID: 37650225 PMCID: PMC10510401 DOI: 10.1021/acs.jmedchem.3c00770] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Indexed: 09/01/2023]
Abstract
NADPH oxidases (NOXs) form a family of electron-transporting membrane enzymes whose main function is reactive oxygen species (ROS) generation. Strong evidence suggests that ROS produced by NOX enzymes are major contributors to oxidative damage under pathologic conditions. Therefore, blocking the undesirable actions of these enzymes is a therapeutic strategy for treating various pathological disorders, such as cardiovascular diseases, inflammation, and cancer. To date, identification of selective NOX inhibitors is quite challenging, precluding a pharmacologic demonstration of NOX as therapeutic targets in vivo. The aim of this Perspective is to furnish an updated outlook about the small-molecule NOX inhibitors described over the last two decades. Structures, activities, and in vitro/in vivo specificity are discussed, as well as the main biological assays used.
Collapse
Affiliation(s)
- Alessandra Cipriano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Monica Viviano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Alessandra Feoli
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Ciro Milite
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Giuliana Sarno
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Sabrina Castellano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Gianluca Sbardella
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| |
Collapse
|
48
|
Chen J, Huang Y, Liu C, Chi J, Wang Y, Xu L. The role of C-peptide in diabetes and its complications: an updated review. Front Endocrinol (Lausanne) 2023; 14:1256093. [PMID: 37745697 PMCID: PMC10512826 DOI: 10.3389/fendo.2023.1256093] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/18/2023] [Indexed: 09/26/2023] Open
Abstract
Worldwide, diabetes and its complications have seriously affected people's quality of life and become a serious public health problem. C-peptide is not only an indicator of pancreatic β-cell function, but also a biologically active peptide that can bind to cell membrane surface signaling molecules and activate downstream signaling pathways to play antioxidant, anti-apoptotic and inflammatory roles, or regulate cellular transcription through internalization. It is complex how C-peptide is related to diabetic complications. Both deficiencies and overproduction can lead to complications, but their mechanisms of action may be different. C-peptide replacement therapy has shown beneficial effects on diabetic complications in animal models when C-peptide is deficient, but results from clinical trials have been unsatisfactory. The complex pattern of the relationship between C-peptide and diabetic chronic complications has not yet been fully understood. Future basic and clinical studies of C-peptide replacement therapies will need to focus on baseline levels of C-peptide in addition to more attention also needs to be paid to post-treatment C-peptide levels to explore the optimal range of fasting C-peptide and postprandial C-peptide maintenance.
Collapse
Affiliation(s)
| | | | | | | | - Yangang Wang
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lili Xu
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
49
|
Yoshida YG, Yan S, Xu H, Yang J. Novel Metal Nanomaterials to Promote Angiogenesis in Tissue Regeneration. ENGINEERED REGENERATION 2023; 4:265-276. [PMID: 37234753 PMCID: PMC10207714 DOI: 10.1016/j.engreg.2023.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Angiogenesis-the formation of new blood vessels from existing blood vessels-has drawn significant attention in medical research. New techniques have been developed to control proangiogenic factors to obtain desired effects. Two important research areas are 1) understanding cellular mechanisms and signaling pathways involved in angiogenesis and 2) discovering new biomaterials and nanomaterials with proangiogenic effects. This paper reviews recent developments in controlling angiogenesis in the context of regenerative medicine and wound healing. We focus on novel proangiogenic materials that will advance the field of regenerative medicine. Specifically, we mainly focus on metal nanomaterials. We also discuss novel technologies developed to carry these proangiogenic inorganic molecules efficiently to target sites. We offer a comprehensive overview by combining existing knowledge regarding metal nanomaterials with novel developments that are still being refined to identify new nanomaterials.
Collapse
Affiliation(s)
- Yuki G. Yoshida
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Su Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Hui Xu
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jian Yang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
50
|
Ribeiro E, Vale N. Understanding the Clinical Use of Levosimendan and Perspectives on its Future in Oncology. Biomolecules 2023; 13:1296. [PMID: 37759695 PMCID: PMC10526140 DOI: 10.3390/biom13091296] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Drug repurposing, also known as repositioning or reprofiling, has emerged as a promising strategy to accelerate drug discovery and development. This approach involves identifying new medical indications for existing approved drugs, harnessing the extensive knowledge of their bioavailability, pharmacokinetics, safety and efficacy. Levosimendan, a calcium sensitizer initially approved for heart failure, has been repurposed for oncology due to its multifaceted pharmacodynamics, including phosphodiesterase 3 inhibition, nitric oxide production and reduction of reactive oxygen species. Studies have demonstrated that levosimendan inhibits cancer cell migration and sensitizes hypoxic cells to radiation. Moreover, it exerts organ-protective effects by activating mitochondrial potassium channels. Combining levosimendan with traditional anticancer agents such as 5-fluorouracil (5-FU) has shown a synergistic effect in bladder cancer cells, highlighting its potential as a novel therapeutic approach. This drug repurposing strategy offers a cost-effective and time-efficient solution for developing new treatments, ultimately contributing to the advancement of cancer therapeutics and improved outcomes for patients. Further investigations and clinical trials are warranted to validate the effectiveness of levosimendan in oncology and explore its potential benefits in a clinical setting.
Collapse
Affiliation(s)
- Eduarda Ribeiro
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|