1
|
Ren C, Shi Z, Zhang X, Yu X, Gao Y, Qi Z, Chen Y, Wang Y. DNA-mediated self-assembly oxidative damage amplifier combined with copper and MTH1 inhibitor for cancer therapy. Bioact Mater 2025; 45:434-445. [PMID: 39697239 PMCID: PMC11653152 DOI: 10.1016/j.bioactmat.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024] Open
Abstract
Chemo-dynamic therapy (CDT) has a great potential in tumor extirpation. It entails producing hypertoxic reactive oxygen species (ROS) that damage the DNA of tumor cells and other biomacromolecules. However, the efficiency of CDT is severely hampered by the massive presence of glutathione (GSH) in tumor cells and the interference of ROS defense systems, such as Mutt homolog 1 (MTH1) protein sanitizes ROS-oxidized nucleotide pools. In this research, DNA-mediated self-assembly nanoparticles (HTCG@TA NPs) were engineered with high-performance amplified oxidative damage and gene therapy effect for synergistic anti-tumor treatment. Cu2+ was converted into Cu + by redox reactions to deplete GSH while H2O2 was catalyzed to generate hydroxyl radicals (·OH). As a result, the ROS level was evidently improved. Moreover, controllable-released TH588 prevented MTH1-mediated DNA repairing, thus aggravated oxidative damage to tumor cells. Meanwhile, the released functional nucleic acid G3139 downregulated the expression of Bcl-2, and accelerated the apoptosis of tumor cells. In conclusion, the HTCG@TA demonstrated significant effect in oxidative damage amplification and tumor inhibition both in vitro and in vivo, which has provided a new outlook for the clinical application of chemo-dynamic tumor treatment and synergistic gene therapy with self-delivery nanoplatforms.
Collapse
Affiliation(s)
- Cui Ren
- Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China
| | - Zhiyong Shi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xiaowen Zhang
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Xueer Yu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yang Gao
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhi Qi
- Department of Molecular Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yu Chen
- Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China
| | - Yong Wang
- Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China
| |
Collapse
|
2
|
Gordillo GM, Guda PR, Singh K, Biswas A, Abouhashem AS, Rustagi Y, Sen A, Kumar M, Das A, Ghatak S, Khanna S, Sen CK, Roy S. Tissue nanotransfection causes tumor regression by its effect on nanovesicle cargo that alters microenvironmental macrophage state. Mol Ther 2023; 31:1402-1417. [PMID: 36380587 PMCID: PMC10188642 DOI: 10.1016/j.ymthe.2022.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 10/25/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) are nanovesicles released by all eukaryotic cells. This work reports the first nanoscale fluorescent visualization of tumor-originating vesicles bearing an angiogenic microRNA (miR)-126 cargo. In a validated experimental model of lethal murine vascular neoplasm, tumor-originating EV delivered its miR-126 cargo to tumor-associated macrophages (TAMs). Such delivery resulted in an angiogenic (LYVE+) change of state in TAM that supported tumor formation. Study of the trafficking of tumor-originating fluorescently tagged EV revealed colocalization with TAM demonstrating uptake by these cells. Ex vivo treatment of macrophages with tumor-derived EVs led to gain of tumorigenicity in these isolated cells. Single-cell RNA sequencing of macrophages revealed that EV-borne miR-126 characterized the angiogenic change of state. Unique gene expression signatures of specific macrophage clusters responsive to miR-126-enriched tumor-derived EVs were revealed. Topical tissue nanotransfection (TNT) delivery of an oligonucleotide comprising an anti-miR against miR-126 resulted in significant knockdown of miR-126 in the tumor tissue. miR-126 knockdown resulted in complete involution of the tumor and improved survival rate of tumor-affected mice. This work identifies a novel tumorigenic mechanism that relies on tumorigenic state change of TAM caused by tumor-originating EV-borne angiomiR. This disease process can be effectively targeted by topical TNT of superficial tumors.
Collapse
Affiliation(s)
- Gayle M Gordillo
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA.
| | - Poornachander Reddy Guda
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Kanhaiya Singh
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Ayan Biswas
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Ahmed S Abouhashem
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Yashika Rustagi
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Abhishek Sen
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Manishekhar Kumar
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Amitava Das
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Subhadip Ghatak
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Savita Khanna
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine and Engineering, Department of Surgery, Indiana University School of Medicine, 975 W Walnut Street, Suite 444, Indianapolis, IN 46202, USA.
| |
Collapse
|
3
|
Miyano K, Okamoto S, Yamauchi A, Kawai C, Kajikawa M, Kiyohara T, Itsumi M, Taura M, Kuribayashi F. The downregulation of NADPH oxidase Nox4 during hypoxia in hemangioendothelioma cells: a possible role of p22 phox on Nox4 protein stability. Free Radic Res 2022; 55:996-1004. [PMID: 35012414 DOI: 10.1080/10715762.2021.2009116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
NADPH oxidase (Nox) 4 produces H2O2 by forming a heterodimer with p22phox and is involved in hemangioendothelioma development through monocyte chemoattractant protein-1 (MCP-1) upregulation. Here, we show that Nox4 protein levels were maintained by p22phox in hemangioendothelioma cells and Nox4 protein stability was dependent on p22phox coexpression. Conversely, the degradation of Nox4 monomer was enhanced by p22phox knockdown. Under hypoxic conditions in hemangioendothelioma cells, p22phox was downregulated at the mRNA and protein levels. Downregulation of p22phox protein resulted in the enhanced degradation of Nox4 protein in hypoxia-treated hemangioendothelioma cells. In contrast, Nox2, a Nox isoform, was not altered at the protein level under hypoxic conditions. Nox2 exhibited a higher affinity for p22phox compared with Nox4, suggesting that when coexpressed with Nox4 in the same cells, Nox2 acts as a competitor. Nox2 knockdown restored Nox4 protein levels partially reduced by hypoxic treatment. Thus, Nox4 protein levels were attenuated in hypoxia-treated cells resulting from p22phox depletion. MCP-1 secretion was decreased concurrently with hypoxia-induced Nox4 downregulation compared with that under normoxia.
Collapse
Affiliation(s)
- Kei Miyano
- Department of Biochemistry, Kawasaki Medical School, Okayama, Japan
| | | | - Akira Yamauchi
- Department of Biochemistry, Kawasaki Medical School, Okayama, Japan
| | - Chikage Kawai
- Department of Biochemistry, Kawasaki Medical School, Okayama, Japan
| | - Mizuho Kajikawa
- Laboratory of Microbiology, Showa Pharmaceutical University, Machida, Japan
| | - Takuya Kiyohara
- Department of Cerebrovascular Disease and Neurology, Hakujyuji Hospital, Fukuoka, Japan.,Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Momoe Itsumi
- Department of Oral Microbiology and Immunology, Showa University, Shinagawa, Japan
| | - Masahiko Taura
- Department of Otorhinolaryngology, Faculty of Medicine, Fukuoka University, Fukuoka City, Japan
| | | |
Collapse
|
4
|
Li XY, Deng FA, Zheng RR, Liu LS, Liu YB, Kong RJ, Chen AL, Yu XY, Li SY, Cheng H. Carrier Free Photodynamic Synergists for Oxidative Damage Amplified Tumor Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102470. [PMID: 34480417 DOI: 10.1002/smll.202102470] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/26/2021] [Indexed: 06/13/2023]
Abstract
Tumor cells adapt to excessive oxidative stress by actuating reactive oxygen species (ROS)-defensing system, leading to a resistance to oxidation therapy. In this work, self-delivery photodynamic synergists (designated as PhotoSyn) are developed for oxidative damage amplified tumor therapy. Specifically, PhotoSyn are fabricated by the self-assembly of chlorine e6 (Ce6) and TH588 through π-π stacking and hydrophobic interactions. Without additional carriers, nanoscale PhotoSyn possess an extremely high drug loading rate (up to 100%) and they are found to be fairly stable in aqueous phase with a uniform size distribution. Intravenously injected PhotoSyn prefer to accumulate at tumor sites for effective cellular uptake. More importantly, TH588-mediated MTH1 inhibition could destroy the ROS-defensing system of tumor cells by preventing the elimination of 8-oxo-2'-deoxyguanosine triphosphate (8-oxo-dG), thereby exacerbating the oxidative DNA damage induced by the photodynamic therapy (PDT) of Ce6 under light irradiation. As a consequence, PhotoSyn exhibit enhanced photo toxicity and a significant antitumor effect. This amplified oxidative damage strategy improves the PDT efficiency with a reduced side effect by increasing the lethality of ROS without generating superabundant ROS, which would provide a new insight for developing self-delivery nanoplatforms in photodynamic tumor therapy in clinic.
Collapse
Affiliation(s)
- Xin-Yu Li
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Fu-An Deng
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Rong-Rong Zheng
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Ling-Shan Liu
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - Yi-Bin Liu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Ren-Jiang Kong
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| | - A-Li Chen
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Xi-Yong Yu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Shi-Ying Li
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, P. R. China
| | - Hong Cheng
- School of Biomedical Engineering & Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, 510515, P. R. China
| |
Collapse
|
5
|
Giorgio M, Dellino GI, Gambino V, Roda N, Pelicci PG. On the epigenetic role of guanosine oxidation. Redox Biol 2020; 29:101398. [PMID: 31926624 PMCID: PMC6926346 DOI: 10.1016/j.redox.2019.101398] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/23/2019] [Accepted: 12/02/2019] [Indexed: 01/14/2023] Open
Abstract
Chemical modifications of DNA and RNA regulate genome functions or trigger mutagenesis resulting in aging or cancer. Oxidations of macromolecules, including DNA, are common reactions in biological systems and often part of regulatory circuits rather than accidental events. DNA alterations are particularly relevant since the unique role of nuclear and mitochondrial genome is coding enduring and inheritable information. Therefore, an alteration in DNA may represent a relevant problem given its transmission to daughter cells. At the same time, the regulation of gene expression allows cells to continuously adapt to the environmental changes that occur throughout the life of the organism to ultimately maintain cellular homeostasis. Here we review the multiple ways that lead to DNA oxidation and the regulation of mechanisms activated by cells to repair this damage. Moreover, we present the recent evidence suggesting that DNA damage caused by physiological metabolism acts as epigenetic signal for regulation of gene expression. In particular, the predisposition of guanine to oxidation might reflect an adaptation to improve the genome plasticity to redox changes.
Collapse
Affiliation(s)
- Marco Giorgio
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy; Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padova, Italy.
| | - Gaetano Ivan Dellino
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Valentina Gambino
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy
| | - Niccolo' Roda
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, Via Adamello 16, 20139, Milano, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
6
|
Du S, Miao J, Lu X, Shi L, Sun J, Xu E, Wang X, Zhao M, Chen H, Wang F, Kang X, Ding J, Guan W, Xia X. NADPH oxidase 4 is correlated with gastric cancer progression and predicts a poor prognosis. Am J Transl Res 2019; 11:3518-3530. [PMID: 31312363 PMCID: PMC6614607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/09/2019] [Indexed: 06/10/2023]
Abstract
NADPH oxidase 4 (NOX4) is one of the main sources of reactive oxygen species, and plays a crucial role in the occurrence and development of tumors. However, there is currently little evidence demonstrating that NOX4 expression is associated with gastric cancer. To establish whether NOX4 plays a role in gastric cancer progression and prognosis, we performed immunohistochemistry on gastric cancer tissues and paired adjacent normal tissues from 90 gastric cancer patients to detect and compare NOX4 expression. Next, we analyzed the association between NOX4 expression and clinicopathological characteristics. Survival analysis was performed to explore the association between NOX4 expression and the prognosis of gastric cancer patients. Furtherly, we investigated the effect of NOX4-knockdown using siRNA on gastric cancer progression in vitro and in vivo. Our results revealed that NOX4 expression in gastric cancer tissues is higher than in paired adjacent normal tissues (P = 0.0009). NOX4 expression is significantly correlated with tumor size (P = 0.0321), lymphatic metastasis (P = 0.0125) and vascular invasion (P = 0.0017) and a poor prognosis (P = 0.0000) in gastric cancer patients. NOX4 depletion could significantly inhibit the invasion, proliferation, EMT and MMP7 expression of gastric cancer cells and suppress the progression of gastric cancer in vivo. In conclusion, NOX4 is related to gastric cancer development and predicts a poor prognosis. NOX4 may play an essential role in the progression of gastric cancer, and is a promising target for the prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Shangce Du
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Ji Miao
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Xiaofeng Lu
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Linsen Shi
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Jie Sun
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - En Xu
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Xingzhou Wang
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Min Zhao
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Hong Chen
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Feng Wang
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Xin Kang
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Jie Ding
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Wenxian Guan
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| | - Xuefeng Xia
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical UniversityGulou District, Nanjing 210008, Jiangsu Province, P. R. China
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School321 Zhongshan Road, Nanjing 210008, Jiangsu Province, P. R. China
| |
Collapse
|
7
|
Kundumani-Sridharan V, Subramani J, Raghavan S, Maiti GP, Owens C, Walker T, Wasnick J, Idell S, Das KC. Short-duration hyperoxia causes genotoxicity in mouse lungs: protection by volatile anesthetic isoflurane. Am J Physiol Lung Cell Mol Physiol 2019; 316:L903-L917. [PMID: 30810065 DOI: 10.1152/ajplung.00142.2018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
High concentrations of oxygen (hyperoxia) are routinely used during anesthesia, and supplemental oxygen is also administered in connection with several other clinical conditions. Although prolonged hyperoxia is known to cause acute lung injury (ALI), whether short-duration hyperoxia causes lung toxicity remains unknown. We exposed mice to room air (RA or 21% O2) or 60% oxygen alone or in combination with 2% isoflurane for 2 h and determined the expression of oxidative stress marker genes, DNA damage and DNA repair genes, and expression of cell cycle regulatory proteins using quantitative PCR and Western analyses. Furthermore, we determined cellular apoptosis using TUNEL assay and assessed the DNA damage product 8-hydroxy-2'-deoxyguanosine (8-Oxo-dG) in the urine of 60% hyperoxia-exposed mice. Our study demonstrates that short-duration hyperoxia causes mitochondrial and nuclear DNA damage and that isoflurane abrogates this DNA damage and decreases apoptosis when used in conjunction with hyperoxia. In contrast, isoflurane mixed with RA caused significant 8-Oxo-dG accumulations in the mitochondria and nucleus. We further show that whereas NADPH oxidase is a major source of superoxide anion generated by isoflurane in normoxia, isoflurane inhibits superoxide generation in hyperoxia. Additionally, isoflurane also protected the mouse lungs against ALI (95% O2 for 36-h exposure). Our study established that short-duration hyperoxia causes genotoxicity in the lungs, which is abrogated when hyperoxia is used in conjunction with isoflurane, but isoflurane alone causes genotoxicity in the lung when delivered with ambient air.
Collapse
Affiliation(s)
| | - Jaganathan Subramani
- Department of Internal Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Somasundaram Raghavan
- Department of Internal Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Guru P Maiti
- Oklahoma Medical Research Foundation , Oklahoma City, Oklahoma
| | - Cade Owens
- Department of Anesthesiology, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Trevor Walker
- Department of Anesthesiology, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - John Wasnick
- Department of Anesthesiology, Texas Tech University Health Sciences Center , Lubbock, Texas
| | - Steven Idell
- Department of Cellular and Molecular Biology, Texas Lung Injury Institute, The University of Texas Health Science Center at Tyler , Tyler, Texas
| | - Kumuda C Das
- Department of Internal Medicine, Texas Tech University Health Sciences Center , Lubbock, Texas
| |
Collapse
|
8
|
YENER Y, YERLİKAYA FH. Western diet induces endogen oxidative deoxyribonucleic acid damage and infl ammation in Wistar rats. REV NUTR 2018. [DOI: 10.1590/1678-98652018000300001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
ABSTRACT Objective Nutritional diseases such as metabolic syndrome, cardiovascular disorder, chronic inflammation or even cancer are observed in people who sustain their lifestyle by Western diet due to high calorie intake. The origin of these diseases are the degraded deoxyribonucleic acid structure. In this study, we investigated whether Western diet produced endogenous oxidative deoxyribonucleic acid damage, apoptosis or inflammation. Methods Twenty-eight male Wistar rats, aged 10-12 weeks, were divided into four groups. The rats in control group received the standard diet and the remaining rats were given one of the following three diets for four weeks: a high-fat diet containing 35% fat, a high-sucrose diet containing 69% sucrose and Western diet comprising both two types of diets. After treatment the serum 8-hydroxy-2-deoxyguanosine, poly (adenosine diphosphate ribose) polymerase-1, chitinase-3-like protein 1, soluble urokinase-type plasminogen activator receptor, Fas ligand and cytochrome c levels were measured. Results It was observed no changes in the serum soluble urokinase-type plasminogen activator receptor, Fas ligand and cytochrome c levels whereas a statistically significant increase in the serum 8-hydroxy-2-deoxyguanosine, poly (adenosine diphosphate ribose) polymerase-1 and chitinase-3-like protein 1 levels were found only in rats that were given Western diet. Conclusion The findings show that Western diet produced endogenous oxidative deoxyribonucleic acid damage, which then increased serum poly (adenosine diphosphate ribose) polymerase-1 levels, eventually leading to inflammation.
Collapse
|
9
|
Moloney JN, Jayavelu AK, Stanicka J, Roche SL, O'Brien RL, Scholl S, Böhmer FD, Cotter TG. Nuclear membrane-localised NOX4D generates pro-survival ROS in FLT3-ITD-expressing AML. Oncotarget 2017; 8:105440-105457. [PMID: 29285262 PMCID: PMC5739649 DOI: 10.18632/oncotarget.22241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/02/2017] [Indexed: 12/21/2022] Open
Abstract
Internal tandem duplication of the juxtamembrane domain of FMS-like tyrosine kinase 3 (FLT3-ITD) is the most prevalent genetic aberration present in 20-30% of acute myeloid leukaemia (AML) cases and is associated with a poor prognosis. FLT3-ITD expressing cells express elevated levels of NADPH oxidase 4 (NOX4)-generated pro-survival hydrogen peroxide (H2O2) contributing to increased levels of DNA oxidation and double strand breaks. NOX4 is constitutively active and has been found to have various isoforms expressed at multiple locations within a cell. The purpose of this study was to investigate the expression, localisation and regulation of NOX4 28 kDa splice variant, NOX4D. NOX4D has previously been shown to localise to the nucleus and nucleolus in various cell types and is implicated in the generation of reactive oxygen species (ROS) and DNA damage. Here, we demonstrate that FLT3-ITD expressing-AML patient samples as well as -cell lines express the NOX4D isoform resulting in elevated H2O2 levels compared to FLT3-WT expressing cells, as quantified by flow cytometry. Cell fractionation indicated that NOX4D is nuclear membrane-localised in FLT3-ITD expressing cells. Treatment of MV4-11 cells with receptor trafficking inhibitors, tunicamycin and brefeldin A, resulted in deglycosylation of NOX4 and NOX4D. Inhibition of the FLT3 receptor revealed that the FLT3-ITD oncogene is responsible for the production of NOX4D-generated H2O2 in AML. We found that inhibition of the PI3K/AKT and STAT5 pathways resulted in down-regulation of NOX4D-generated pro-survival ROS. Taken together these findings indicate that nuclear membrane-localised NOX4D-generated pro-survival H2O2 may be contributing to genetic instability in FLT3-ITD expressing AML.
Collapse
Affiliation(s)
- Jennifer N Moloney
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Ashok Kumar Jayavelu
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany.,Current address: Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Joanna Stanicka
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Sarah L Roche
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Rebecca L O'Brien
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| | - Sebastian Scholl
- Department of Haematology/Oncology, Clinic for Internal Medicine II, Jena University Hospital, Jena, Germany
| | - Frank-D Böhmer
- Institute of Molecular Cell Biology, CMB, Jena University Hospital, Jena, Germany
| | - Thomas G Cotter
- Tumour Biology Laboratory, School of Biochemistry and Cell Biology, Bioscience Research Institute, University College Cork, Cork, Ireland
| |
Collapse
|
10
|
Reply: Urinary Excretion of MicroRNA-126 Is a Biomarker for Hemangioma Proliferation. Plast Reconstr Surg 2017; 141:320e. [PMID: 29068892 DOI: 10.1097/prs.0000000000004090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Gu R, Sun X, Chi Y, Zhou Q, Xiang H, Bosco DB, Lai X, Qin C, So KF, Ren Y, Chen XM. Integrin β3/Akt signaling contributes to platelet-induced hemangioendothelioma growth. Sci Rep 2017; 7:6455. [PMID: 28744026 PMCID: PMC5527091 DOI: 10.1038/s41598-017-06927-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 06/22/2017] [Indexed: 01/16/2023] Open
Abstract
Hemangioendothelioma (HE) is a type of angiomatous lesions that features endothelial cell proliferation. Understanding the mechanisms orchestrating HE angiogenesis can provide therapeutic insights. It has been shown that platelets can support normal and malignant endothelial cells during angiogenesis. Using the mouse endothelial-derived EOMA cell line as a model of HE, we explored the regulatory effect of platelets. We found that platelets stimulated EOMA proliferation but did not mitigate apoptosis. Furthermore, direct platelet-EOMA cell contact was required and the proliferation was mediated via integrin β3/Akt signaling in EOMA cells. SiRNA knockdown of integrin β3 and inhibition of Akt activity significantly abolished platelet-induced EOMA cell proliferation in vitro and tumor development in vivo. These results provide a new mechanism by which platelets support HE progression and suggest integrin β3 as a potential target to treat HE.
Collapse
Affiliation(s)
- Rui Gu
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Sun
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Joint International Research Laboratory of CNS Regeneration Ministry of Education, Guangdong Medical Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong, China
| | - Yijie Chi
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qishuang Zhou
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongkai Xiang
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dale B Bosco
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Xinhe Lai
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Caixia Qin
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Joint International Research Laboratory of CNS Regeneration Ministry of Education, Guangdong Medical Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Joint International Research Laboratory of CNS Regeneration Ministry of Education, Guangdong Medical Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong, China
| | - Yi Ren
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China. .,Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Joint International Research Laboratory of CNS Regeneration Ministry of Education, Guangdong Medical Key Laboratory of Brain Function and Diseases, Jinan University, Guangzhou, China. .,Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA.
| | - Xiao-Ming Chen
- Institute of Inflammation and Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
12
|
Biswas A, Clark EC, Sen CK, Gordillo GM. Phytochemical Inhibition of Multidrug Resistance Protein-1 as a Therapeutic Strategy for Hemangioendothelioma. Antioxid Redox Signal 2017; 26:1009-1019. [PMID: 27706944 PMCID: PMC5467139 DOI: 10.1089/ars.2016.6881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIMS Hemangiomas are endothelial cell tumors and the most common soft tissue tumors in infants. They frequently cause deformity and can cause death. Current pharmacologic therapies have high-risk side-effect profiles, which limit the number of children who receive treatment. The objectives of this work were to identify the mechanisms through which standardized berry extracts can inhibit endothelial cell tumor growth and test these findings in vivo. RESULTS EOMA cells are a validated model that generates endothelial cell tumors when injected subcutaneously into syngeneic (129P/3) mice. EOMA cells treated with a blend of powdered natural berry extracts (NBE) significantly inhibited activity of multidrug resistance protein-1 (MRP-1) compared to vehicle controls. This resulted in nuclear accumulation of oxidized glutathione (GSSG) and apoptotic EOMA cell death. When NBE-treated EOMA cells were injected into mice, they generated smaller tumors and had a higher incidence of apoptotic cell death compared to vehicle-treated EOMA cells as demonstrated by immunocytochemistry. Kaplan-Meier survival curves for tumor-bearing mice showed that NBE treatment significantly prolonged survival compared to vehicle-treated controls. INNOVATION These are the first reported results to show that berry extracts can inhibit MRP-1 function that causes apoptotic tumor cell death by accumulation of GSSG in the nucleus of EOMA cells where NADPH oxidase is hyperactive and causes pathological angiogenesis. CONCLUSIONS These findings indicate that berry extract inhibition of MRP-1 merits consideration and further investigation as a therapeutic intervention and may have application for other cancers with elevated MRP-1 activity. Antioxid. Redox Signal. 26, 1009-1019.
Collapse
Affiliation(s)
- Ayan Biswas
- 1 Department of Plastic Surgery, The Ohio State University , Columbus, Ohio
| | - Emma C Clark
- 1 Department of Plastic Surgery, The Ohio State University , Columbus, Ohio
| | - Chandan K Sen
- 2 Department of Surgery, David Heart and Lung Research Institute, The Ohio State University , Columbus, Ohio
| | - Gayle M Gordillo
- 1 Department of Plastic Surgery, The Ohio State University , Columbus, Ohio
| |
Collapse
|
13
|
Biswas A, Pan X, Meyer M, Khanna S, Roy S, Pearson G, Kirschner R, Witman P, Faith EF, Sen CK, Gordillo GM. Urinary Excretion of MicroRNA-126 Is a Biomarker for Hemangioma Proliferation. Plast Reconstr Surg 2017; 139:1277e-1284e. [PMID: 28538565 PMCID: PMC5963954 DOI: 10.1097/prs.0000000000003349] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Hemangiomas are unique endothelial cell tumors that involute spontaneously, which makes interpreting their response to therapies difficult. The objective of this work was to identify a potential biomarker in the urine of children with infantile hemangiomas that would facilitate testing new therapies. METHODS A prospective longitudinal study in children with hemangiomas and age-matched healthy controls was performed to determine whether microRNA-126, which is highly abundant in fetal endothelial cells, was more abundant in the urine of affected children. Prospective ultrasound measurements of hemangioma size and blood flow velocity were obtained as secondary endpoints to document longitudinal changes in untreated hemangiomas. RESULTS Urinary microRNA-126 levels were significantly elevated in children with proliferating hemangiomas, and relative levels of urinary microRNA abundance correlated with hemangioma size. Hemangiomas had elevated levels of microRNA abundance compared with healthy controls. Ultrasound data revealed that hemangioma proliferation typically stopped between 6 and 9 months of age. When hemangioma proliferation stopped, urinary microRNA-126 levels in children with hemangiomas dropped to levels observed in healthy age-matched controls. CONCLUSIONS These are the first reported results to identify a potential microRNA biomarker in the urine of children with hemangiomas. Measurement of urinary levels of microRNA-126 could potentially be used to monitor hemangioma response to therapies. CLINICAL QUESTION/LEVEL OF EVIDENCE Diagnostic, II.
Collapse
Affiliation(s)
- Ayan Biswas
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Xueliang Pan
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Melissa Meyer
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Savita Khanna
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Sashwati Roy
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Gregory Pearson
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Richard Kirschner
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Patricia Witman
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Esteban Fernandez Faith
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Chandan K Sen
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| | - Gayle M Gordillo
- Columbus, Ohio
- From the Department of Plastic Surgery, the Department of Biomedical Informatics, Center for Biostatistics, the Department of Surgery, and the Divisions of Vascular Surgery and General Surgery, The Ohio State University; and the Department of Pediatrics, the Hemangioma and Vascular Malformation Clinic, and the Department of Pediatrics, Division of Dermatology, Nationwide Children's Hospital
| |
Collapse
|
14
|
NADPH Oxidases: Insights into Selected Functions and Mechanisms of Action in Cancer and Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017. [PMID: 28626501 PMCID: PMC5463201 DOI: 10.1155/2017/9420539] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
NADPH oxidases (NOX) are reactive oxygen species- (ROS-) generating enzymes regulating numerous redox-dependent signaling pathways. NOX are important regulators of cell differentiation, growth, and proliferation and of mechanisms, important for a wide range of processes from embryonic development, through tissue regeneration to the development and spread of cancer. In this review, we discuss the roles of NOX and NOX-derived ROS in the functioning of stem cells and cancer stem cells and in selected aspects of cancer cell physiology. Understanding the functions and complex activities of NOX is important for the application of stem cells in tissue engineering, regenerative medicine, and development of new therapies toward invasive forms of cancers.
Collapse
|
15
|
Panth N, Paudel KR, Parajuli K. Reactive Oxygen Species: A Key Hallmark of Cardiovascular Disease. Adv Med 2016; 2016:9152732. [PMID: 27774507 PMCID: PMC5059509 DOI: 10.1155/2016/9152732] [Citation(s) in RCA: 227] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/11/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) have been the prime cause of mortality worldwide for decades. However, the underlying mechanism of their pathogenesis is not fully clear yet. It has been already established that reactive oxygen species (ROS) play a vital role in the progression of CVDs. ROS are chemically unstable reactive free radicals containing oxygen, normally produced by xanthine oxidase, nicotinamide adenine dinucleotide phosphate oxidase, lipoxygenases, or mitochondria or due to the uncoupling of nitric oxide synthase in vascular cells. When the equilibrium between production of free radicals and antioxidant capacity of human physiology gets altered due to several pathophysiological conditions, oxidative stress is induced, which in turn leads to tissue injury. This review focuses on pathways behind the production of ROS, its involvement in various intracellular signaling cascades leading to several cardiovascular disorders (endothelial dysfunction, ischemia-reperfusion, and atherosclerosis), methods for its detection, and therapeutic strategies for treatment of CVDs targeting the sources of ROS. The information generated by this review aims to provide updated insights into the understanding of the mechanisms behind cardiovascular complications mediated by ROS.
Collapse
Affiliation(s)
- Nisha Panth
- Department of Pharmacy, School of Health and Allied Sciences, Pokhara University, Dhungepatan, Kaski 33701, Nepal
| | - Keshav Raj Paudel
- Department of Pharmacy, School of Health and Allied Sciences, Pokhara University, Dhungepatan, Kaski 33701, Nepal
| | - Kalpana Parajuli
- Department of Pharmacy, School of Health and Allied Sciences, Pokhara University, Dhungepatan, Kaski 33701, Nepal
| |
Collapse
|
16
|
Gordillo GM, Biswas A, Khanna S, Spieldenner JM, Pan X, Sen CK. Multidrug Resistance-associated Protein-1 (MRP-1)-dependent Glutathione Disulfide (GSSG) Efflux as a Critical Survival Factor for Oxidant-enriched Tumorigenic Endothelial Cells. J Biol Chem 2016; 291:10089-103. [PMID: 26961872 DOI: 10.1074/jbc.m115.688879] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Indexed: 12/28/2022] Open
Abstract
Endothelial cell tumors are the most common soft tissue tumors in infants. Tumor-forming endothelial (EOMA) cells are able to escape cell death fate despite excessive nuclear oxidant burden. Our previous work recognized perinuclear Nox-4 as a key contributor to EOMA growth. The objective of this work was to characterize the mechanisms by which EOMA cells evade oxidant toxicity and thrive. In EOMA cells, compared with in the cytosol, the nuclear GSSG/GSH ratio was 5-fold higher. Compared to the ratio observed in healthy murine aortic endothelial (MAE) cells, GSSG/GSH was over twice as high in EOMA cells. Multidrug resistance-associated protein-1 (MRP-1), an active GSSG efflux mechanism, showed 2-fold increased activity in EOMA compared with MAE cells. Hyperactive YB-1 and Ape/Ref-1 were responsible for high MRP-1 expression in EOMA. Proximity ligand assay demonstrated MRP-1 and YB-1 binding. Such binding enabled the nuclear targeting of MRP-1 in EOMA in a leptomycin-B-sensitive manner. MRP-1 inhibition as well as knockdown trapped nuclear GSSG, causing cell death of EOMA. Disulfide loading of cells by inhibition of GSSG reductase (bischoloronitrosourea) or thioredoxin reductase (auranofin) was effective in causing EOMA death as well. In sum, EOMA cells survive a heavy oxidant burden by rapid efflux of GSSG, which is lethal if trapped within the cell. A hyperactive MRP-1 system for GSSG efflux acts as a critical survival factor for these cells, making it a potential target for EOMA therapeutics.
Collapse
Affiliation(s)
- Gayle M Gordillo
- From the Department of Plastic Surgery, Davis Heart and Lung Research Institute, and
| | - Ayan Biswas
- From the Department of Plastic Surgery, Davis Heart and Lung Research Institute, and
| | - Savita Khanna
- Davis Heart and Lung Research Institute, and Department of Surgery
| | | | - Xueliang Pan
- Center for Biostatistics, Ohio State University Wexner Medical Center, Columbus, Ohio 43212
| | - Chandan K Sen
- Davis Heart and Lung Research Institute, and Department of Surgery
| |
Collapse
|
17
|
Kim GH, Kim JE, Rhie SJ, Yoon S. The Role of Oxidative Stress in Neurodegenerative Diseases. Exp Neurobiol 2015; 24:325-40. [PMID: 26713080 PMCID: PMC4688332 DOI: 10.5607/en.2015.24.4.325] [Citation(s) in RCA: 920] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 09/14/2015] [Accepted: 09/14/2015] [Indexed: 12/14/2022] Open
Abstract
Oxidative stress is induced by an imbalanced redox states, involving either excessive generation of reactive oxygen species (ROS) or dysfunction of the antioxidant system. The brain is one of organs especially vulnerable to the effects of ROS because of its high oxygen demand and its abundance of peroxidation-susceptible lipid cells. Previous studies have demonstrated that oxidative stress plays a central role in a common pathophysiology of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Antioxidant therapy has been suggested for the prevention and treatment of neurodegenerative diseases, although the results with regard to their efficacy of treating neurodegenerative disease have been inconsistent. In this review, we will discuss the role of oxidative stress in the pathophysiology of neurodegenerative diseases and in vivo measurement of an index of damage by oxidative stress. Moreover, the present knowledge on antioxidant in the treatment of neurodegenerative diseases and future directions will be outlined.
Collapse
Affiliation(s)
- Geon Ha Kim
- Ewha Brain Institute, Ewha Womans University, Seoul 03760, Korea. ; Department of Neurology, Ewha Womans University Mokdong Hospital, Ewha Womans University School of Medicine, Seoul 03760, Korea
| | - Jieun E Kim
- Ewha Brain Institute, Ewha Womans University, Seoul 03760, Korea. ; Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Sandy Jeong Rhie
- Ewha Brain Institute, Ewha Womans University, Seoul 03760, Korea. ; College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Sujung Yoon
- Ewha Brain Institute, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
18
|
Biswas A, Khanna S, Roy S, Pan X, Sen CK, Gordillo GM. Endothelial cell tumor growth is Ape/ref-1 dependent. Am J Physiol Cell Physiol 2015; 309:C296-307. [PMID: 26108661 DOI: 10.1152/ajpcell.00022.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/17/2015] [Indexed: 01/12/2023]
Abstract
Tumor-forming endothelial cells have highly elevated levels of Nox-4 that release H2O2 into the nucleus, which is generally not compatible with cell survival. We sought to identify compensatory mechanisms that enable tumor-forming endothelial cells to survive and proliferate under these conditions. Ape-1/ref-1 (Apex-1) is a multifunctional protein that promotes DNA binding of redox-sensitive transcription factors, such as AP-1, and repairs oxidative DNA damage. A validated mouse endothelial cell (EOMA) tumor model was used to demonstrate that Nox-4-derived H2O2 causes DNA oxidation that induces Apex-1 expression. Apex-1 functions as a chaperone to keep transcription factors in a reduced state. In EOMA cells Apex-1 enables AP-1 binding to the monocyte chemoattractant protein-1 (mcp-1) promoter and expression of that protein is required for endothelial cell tumor formation. Intraperitoneal injection of the small molecule inhibitor E3330, which specifically targets Apex-1 redox-sensitive functions, resulted in a 50% decrease in tumor volume compared with mice injected with vehicle control (n = 6 per group), indicating that endothelial cell tumor proliferation is dependent on Apex-1 expression. These are the first reported results to establish Nox-4 induction of Apex-1 as a mechanism promoting endothelial cell tumor formation.
Collapse
Affiliation(s)
- Ayan Biswas
- Department of Plastic Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Savita Khanna
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Sashwati Roy
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Xueliang Pan
- Center for Biostatistics, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Chandan K Sen
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Gayle M Gordillo
- Department of Plastic Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| |
Collapse
|
19
|
Dessì C, Leoni G, Moi P, Danjou F, Follesa I, Foschini ML, Morittu M, Zappu A, Defraia E, Bina P, Cunico A, Civolani A, Podda RA, Origa R. Thalassemia major between liver and heart: Where we are now. Blood Cells Mol Dis 2015; 55:82-8. [DOI: 10.1016/j.bcmd.2015.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 03/29/2015] [Indexed: 02/08/2023]
|
20
|
|
21
|
Rinna A, Magdolenova Z, Hudecova A, Kruszewski M, Refsnes M, Dusinska M. Effect of silver nanoparticles on mitogen-activated protein kinases activation: role of reactive oxygen species and implication in DNA damage. Mutagenesis 2014; 30:59-66. [DOI: 10.1093/mutage/geu057] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
22
|
McCarty MF, Contreras F. Increasing Superoxide Production and the Labile Iron Pool in Tumor Cells may Sensitize Them to Extracellular Ascorbate. Front Oncol 2014; 4:249. [PMID: 25279352 PMCID: PMC4165285 DOI: 10.3389/fonc.2014.00249] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/01/2014] [Indexed: 12/23/2022] Open
Abstract
Low millimolar concentrations of ascorbate are capable of inflicting lethal damage on a high proportion of cancer cells lines, yet leave non-transformed cell lines unscathed. Extracellular generation of hydrogen peroxide, reflecting reduction of molecular oxygen by ascorbate, has been shown to mediate this effect. Although some cancer cell lines express low catalase activity, this cannot fully explain the selective sensitivity of cancer cells to hydrogen peroxide. Ranzato and colleagues have presented evidence for a plausible new explanation of this sensitivity - a high proportion of cancers, via NADPH oxidase complexes or dysfunctional mitochondria, produce elevated amounts of superoxide. This superoxide, via a transition metal-catalyzed transfer of an electron to the hydrogen peroxide produced by ascorbate, can generate deadly hydroxyl radical (Haber-Weiss reaction). It thus can be predicted that concurrent measures which somewhat selectively boost superoxide production in cancers will enhance their sensitivity to i.v. ascorbate therapy. One way to achieve this is to increase the provision of substrate to cancer mitochondria. Measures which inhibit the constitutive hypoxia-inducible factor-1 (HIF-1) activity in cancers (such as salsalate and mTORC1 inhibitors, or an improvement of tumor oxygenation), or that inhibit the HIF-1-inducible pyruvate dehydrogenase kinase (such as dichloroacetate), can be expected to increase pyruvate oxidation. A ketogenic diet should provide more lipid substrate for tumor mitochondria. The cancer-killing activity of 42°C hyperthermia is to some degree contingent on an increase in oxidative stress, likely of mitochondrial origin; reports that hydrogen peroxide synergizes with hyperthermia in killing cancer cells suggest that hyperthermia and i.v. ascorbate could potentiate each other's efficacy. A concurrent enhancement of tumor oxygenation might improve results by decreasing HIF-1 activity while increasing the interaction of ascorbic acid with oxygen. An increased pool of labile iron in cancer cells may contribute to the selective susceptibility of many cancers to i.v. ascorbate; antagonism of NF-kappaB activity with salicylate, and intravenous iron administration, could be employed to further elevate free iron in cancers.
Collapse
|
23
|
Choi J, Corder NLB, Koduru B, Wang Y. Oxidative stress and hepatic Nox proteins in chronic hepatitis C and hepatocellular carcinoma. Free Radic Biol Med 2014; 72:267-84. [PMID: 24816297 PMCID: PMC4099059 DOI: 10.1016/j.freeradbiomed.2014.04.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Revised: 04/16/2014] [Accepted: 04/18/2014] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common liver cancer and a leading cause of cancer-related mortality in the world. Hepatitis C virus (HCV) is a major etiologic agent of HCC. A majority of HCV infections lead to chronic infection that can progress to cirrhosis and, eventually, HCC and liver failure. A common pathogenic feature present in HCV infection, and other conditions leading to HCC, is oxidative stress. HCV directly increases superoxide and H2O2 formation in hepatocytes by elevating Nox protein expression and sensitizing mitochondria to reactive oxygen species generation while decreasing glutathione. Nitric oxide synthesis and hepatic iron are also elevated. Furthermore, activation of phagocytic NADPH oxidase (Nox) 2 of host immune cells is likely to exacerbate oxidative stress in HCV-infected patients. Key mechanisms of HCC include genome instability, epigenetic regulation, inflammation with chronic tissue injury and sustained cell proliferation, and modulation of cell growth and death. Oxidative stress, or Nox proteins, plays various roles in these mechanisms. Nox proteins also function in hepatic fibrosis, which commonly precedes HCC, and Nox4 elevation by HCV is mediated by transforming growth factor β. This review summarizes mechanisms of oncogenesis by HCV, highlighting the roles of oxidative stress and hepatic Nox enzymes in HCC.
Collapse
Affiliation(s)
- Jinah Choi
- School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA.
| | - Nicole L B Corder
- School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA
| | - Bhargav Koduru
- School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA
| | - Yiyan Wang
- School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA
| |
Collapse
|
24
|
Hou H, Wang X, Chen C, Johnson DM, Fang Y, Huang Y. Mechanism of photocatalytic oxidation of guanine by BiOBr under UV irradiation. CATAL COMMUN 2014. [DOI: 10.1016/j.catcom.2014.01.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
25
|
Gordillo GM, Biswas A, Khanna S, Pan X, Sinha M, Roy S, Sen CK. Dicer knockdown inhibits endothelial cell tumor growth via microRNA 21a-3p targeting of Nox-4. J Biol Chem 2014; 289:9027-38. [PMID: 24497637 DOI: 10.1074/jbc.m113.519264] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
MicroRNAs (miR) are emerging as biomarkers and potential therapeutic targets in tumor management. Endothelial cell tumors are the most common soft tissue tumors in infants, yet little is known about the significance of miR in regulating their growth. A validated mouse endothelial cell (EOMA) tumor model was used to demonstrate that post-transcriptional gene silencing of dicer, the enzyme that converts pre-miR to mature miR, can prevent tumor formation in vivo. Tumors were formed in eight of eight mice injected with EOMA cells transfected with control shRNA but formed in only four of ten mice injected with EOMA cells transfected with dicer shRNA. Tumors that formed in the dicer shRNA group were significantly smaller than tumors in the control group. This response to dicer knockdown was mediated by up-regulated miR 21a-3p activity targeting the nox-4 3'-UTR. EOMA cells were transfected with miR 21a-3p mimic and luciferase reporter plasmids containing either intact nox-4 3'-UTR or with mutation of the proposed 3'-UTR miR21a-3p binding sites. Mean luciferase activity was decreased by 85% in the intact compared with the site mutated vectors (p < 0.01). Attenuated Nox-4 activity resulted in decreased cellular hydrogen peroxide production and decreased production of oxidant-inducible monocyte chemoattractant protein-1, which we have previously shown to be critically required for endothelial cell tumor formation. These findings provide the first evidence establishing the significance of dicer and microRNA in promoting endothelial cell tumor growth in vivo.
Collapse
|
26
|
Inhibition of nuclear Nox4 activity by plumbagin: effect on proliferative capacity in human amniotic stem cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:680816. [PMID: 24489986 PMCID: PMC3893878 DOI: 10.1155/2013/680816] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 11/18/2013] [Accepted: 11/19/2013] [Indexed: 01/03/2023]
Abstract
Human amniotic fluid stem cells (AFSC) with multilineage differentiation potential are novel source for cell therapy. However, in vitro expansion leads to senescence affecting differentiation and proliferative capacities. Reactive oxygen species (ROS) have been involved in the regulation of stem cell pluripotency, proliferation, and differentiation. Redox-regulated signal transduction is coordinated by spatially controlled production of ROS within subcellular compartments. NAD(P)H oxidase family, in particular Nox4, has been known to produce ROS in the nucleus; however, the mechanisms and the meaning of this function remain largely unknown. In the present study, we show that Nox4 nuclear expression (nNox4) increases during culture passages up to cell cycle arrest and the serum starvation causes the same effect. With the decrease of Nox4 activity, obtained with plumbagin, a decline of nuclear ROS production and of DNA damage occurs. Moreover, plumbagin exposure reduces the binding between nNox4 and nucleoskeleton components, as Matrin 3. The same effect was observed also for the binding with phospho-ERK, although nuclear ERK and P-ERK are unchanged. Taken together, we suggest that nNox4 regulation may have important pathophysiologic effects in stem cell proliferation through modulation of nuclear signaling and DNA damage.
Collapse
|
27
|
Nguyen Dinh Cat A, Montezano AC, Burger D, Touyz RM. Angiotensin II, NADPH oxidase, and redox signaling in the vasculature. Antioxid Redox Signal 2013; 19:1110-20. [PMID: 22530599 PMCID: PMC3771549 DOI: 10.1089/ars.2012.4641] [Citation(s) in RCA: 325] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
SIGNIFICANCE Angiotensin II (Ang II) influences the function of many cell types and regulates many organ systems, in large part through redox-sensitive processes. In the vascular system, Ang II is a potent vasoconstrictor and also promotes inflammation, hypertrophy, and fibrosis, which are important in vascular damage and remodeling in cardiovascular diseases. The diverse actions of Ang II are mediated via Ang II type 1 and Ang II type 2 receptors, which couple to various signaling molecules, including NADPH oxidase (Nox), which generates reactive oxygen species (ROS). ROS are now recognized as signaling molecules, critically placed in pathways activated by Ang II. Mechanisms linking Nox and Ang II are complex and not fully understood. RECENT ADVANCES Ang II regulates vascular cell production of ROS through various recently characterized Noxs, including Nox1, Nox2, Nox4, and Nox5. Activation of these Noxs leads to ROS generation, which in turn influences many downstream signaling targets of Ang II, including MAP kinases, RhoA/Rho kinase, transcription factors, protein tyrosine phosphatases, and tyrosine kinases. Activation of these redox-sensitive pathways regulates vascular cell growth, inflammation, contraction, and senescence. CRITICAL ISSUES Although there is much evidence indicating a role for Nox/ROS in Ang II function, there is still a paucity of information on how Ang II exerts cell-specific effects through ROS and how Nox isoforms are differentially regulated by Ang II. Moreover, exact mechanisms whereby ROS induce oxidative modifications of signaling molecules mediating Ang II actions remain elusive. FUTURE DIRECTIONS Future research should elucidate these issues to better understand the significance of Ang II and ROS in vascular (patho) biology.
Collapse
Affiliation(s)
- Aurelie Nguyen Dinh Cat
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
| | - Augusto C. Montezano
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
| | - Dylan Burger
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
| | - Rhian M. Touyz
- Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa, Ontario, Canada
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
28
|
Policastro LL, Ibañez IL, Notcovich C, Duran HA, Podhajcer OL. The tumor microenvironment: characterization, redox considerations, and novel approaches for reactive oxygen species-targeted gene therapy. Antioxid Redox Signal 2013; 19:854-95. [PMID: 22794113 DOI: 10.1089/ars.2011.4367] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The tumor microenvironment is a complex system that involves the interaction between malignant and neighbor stromal cells embedded in a mesh of extracellular matrix (ECM) components. Stromal cells (fibroblasts, endothelial, and inflammatory cells) are co-opted at different stages to help malignant cells invade the surrounding ECM and disseminate. Malignant cells have developed adaptive mechanisms to survive under the extreme conditions of the tumor microenvironment such as restricted oxygen supply (hypoxia), nutrient deprivation, and a prooxidant state among others. These conditions could be eventually used to target drugs that will be activated specifically in this microenvironment. Preclinical studies have shown that modulating cellular/tissue redox state by different gene therapy (GT) approaches was able to control tumor growth. In this review, we describe the most relevant features of the tumor microenvironment, addressing reactive oxygen species-generating sources that promote a prooxidative microenvironment inside the tumor mass. We describe different GT approaches that promote either a decreased or exacerbated prooxidative microenvironment, and those that make use of the differential levels of ROS between cancer and normal cells to achieve tumor growth inhibition.
Collapse
Affiliation(s)
- Lucia Laura Policastro
- Department of Micro and Nanotechnology, National Atomic Energy Commission, Buenos Aires 1650, Argentina.
| | | | | | | | | |
Collapse
|
29
|
Hajas G, Bacsi A, Aguilera-Aguirre L, Hegde ML, Tapas KH, Sur S, Radak Z, Ba X, Boldogh I. 8-Oxoguanine DNA glycosylase-1 links DNA repair to cellular signaling via the activation of the small GTPase Rac1. Free Radic Biol Med 2013; 61:384-94. [PMID: 23612479 PMCID: PMC3795866 DOI: 10.1016/j.freeradbiomed.2013.04.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2012] [Revised: 02/24/2013] [Accepted: 04/09/2013] [Indexed: 12/20/2022]
Abstract
8-Oxo-7,8-dihydroguanine (8-oxoG) is one of the most abundant DNA base lesions induced by reactive oxygen species (ROS). Accumulation of 8-oxoG in the mammalian genome is considered a marker of oxidative stress, to be causally linked to inflammation, and is thought to contribute to aging processes and various aging-related diseases. Unexpectedly, mice that lack 8-oxoguanine DNA glycosylase-1 (OGG1) activity and accumulate 8-oxoG in their genome have a normal phenotype and longevity; in fact, they show increased resistance to both inflammation and oxidative stress. OGG1 excises and generates free 8-oxoG base during DNA base-excision repair (BER) processes. In the present study, we report that in the presence of the 8-oxoG base, OGG1 physically interacts with guanine nucleotide-free and GDP-bound Rac1 protein. This interaction results in rapid GDP→GTP, but not GTP→GDP, exchange in vitro. Importantly, a rise in the intracellular 8-oxoG base levels increases the proportion of GTP-bound Rac1. In turn Rac1-GTP mediates an increase in ROS levels via nuclear membrane-associated NADPH oxidase type 4. These results show a novel mechanism by which OGG1 in complex with 8-oxoG is linked to redox signaling and cellular responses.
Collapse
Affiliation(s)
- Gyorgy Hajas
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Attila Bacsi
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Leopoldo Aguilera-Aguirre
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Muralidhar L Hegde
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - K Hazra Tapas
- Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Department of Biochemistry & Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Sanjiv Sur
- Department of Internal Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Zsolt Radak
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Xueqing Ba
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA; Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| |
Collapse
|
30
|
Rodiño-Janeiro BK, Paradela-Dobarro B, Castiñeiras-Landeira MI, Raposeiras-Roubín S, González-Juanatey JR, Álvarez E. Current status of NADPH oxidase research in cardiovascular pharmacology. Vasc Health Risk Manag 2013; 9:401-28. [PMID: 23983473 PMCID: PMC3750863 DOI: 10.2147/vhrm.s33053] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The implications of reactive oxygen species in cardiovascular disease have been known for some decades. Rationally, therapeutic antioxidant strategies combating oxidative stress have been developed, but the results of clinical trials have not been as good as expected. Therefore, to move forward in the design of new therapeutic strategies for cardiovascular disease based on prevention of production of reactive oxygen species, steps must be taken on two fronts, ie, comprehension of reduction-oxidation signaling pathways and the pathophysiologic roles of reactive oxygen species, and development of new, less toxic, and more selective nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitors, to clarify both the role of each NADPH oxidase isoform and their utility in clinical practice. In this review, we analyze the value of NADPH oxidase as a therapeutic target for cardiovascular disease and the old and new pharmacologic agents or strategies to prevent NADPH oxidase activity. Some inhibitors and different direct or indirect approaches are available. Regarding direct NADPH oxidase inhibition, the specificity of NADPH oxidase is the focus of current investigations, whereas the chemical structure-activity relationship studies of known inhibitors have provided pharmacophore models with which to search for new molecules. From a general point of view, small-molecule inhibitors are preferred because of their hydrosolubility and oral bioavailability. However, other possibilities are not closed, with peptide inhibitors or monoclonal antibodies against NADPH oxidase isoforms continuing to be under investigation as well as the ongoing search for naturally occurring compounds. Likewise, some different approaches include inhibition of assembly of the NADPH oxidase complex, subcellular translocation, post-transductional modifications, calcium entry/release, electron transfer, and genetic expression. High-throughput screens for any of these activities could provide new inhibitors. All this knowledge and the research presently underway will likely result in development of new drugs for inhibition of NADPH oxidase and application of therapeutic approaches based on their action, for the treatment of cardiovascular disease in the next few years.
Collapse
Affiliation(s)
- Bruno K Rodiño-Janeiro
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- European Molecular Biology Laboratory, Grenoble, France
| | | | | | - Sergio Raposeiras-Roubín
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- Cardiology Department, University Clinic Hospital of Santiago de Compostela,
Santiago de Compostela, Spain
| | - José R González-Juanatey
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- Cardiology Department, University Clinic Hospital of Santiago de Compostela,
Santiago de Compostela, Spain
- Medicine Department, University of Santiago de Compostela, Santiago de Compostela,
Spain
| | - Ezequiel Álvarez
- Health Research Institute of Santiago de Compostela, Santiago de Compostela,
Spain
- Medicine Department, University of Santiago de Compostela, Santiago de Compostela,
Spain
| |
Collapse
|
31
|
Weyemi U, Redon CE, Parekh PR, Dupuy C, Bonner WM. NADPH Oxidases NOXs and DUOXs as putative targets for cancer therapy. Anticancer Agents Med Chem 2013; 13:502-514. [PMID: 22931418 PMCID: PMC6365101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2012] [Revised: 07/03/2012] [Accepted: 07/23/2012] [Indexed: 06/01/2023]
Abstract
Reactive oxygen species (ROS) form a class of molecules with both positive and negative impacts on cellular health. Negatively, ROS may react with cellular constituents including proteins, lipids, and DNA to generate an array of oxidative lesions. These lesions may compromise genome stability which is critical for long-term cellular homeostasis and healthy progeny. Paradoxically, ROS also function as strong signalling molecules that mediate various growth-related responses, so their presence is also essential for cellular metabolism. While ROS are generated in an unregulated manner by physical stresses such as exposure to ionizing radiation and biochemical malfunctions such as mitochondrial leakage, cells also contain the NADPH oxidases NOXs and DUOXs, which specifically generate ROS in a wide variety of tissues. While the NOXs/DUOXs may be involved in maintaining optimal cellular redox levels, there is also accumulating evidence that NADPH oxidases-derived ROS may elevate the risk for genomic instability and cancer. Cancer cells may produce high levels of ROS, and in some cases, the source of these ROS has been linked to NOX/DUOX deregulation as reported for prostate cancer (NOX1 and NOX5), melanoma and glioblastoma (NOX4) among others. In addition, recent studies reveal that targeting NADPH oxidases with NOXs inhibitors may impair tumor growth in vivo; indicating that these proteins may be useful targets in future clinical strategies to fight cancer. This review provides an overview of the current knowledge concerning these enzymes, their roles in cancer, and their potential as targets in future cancer therapies.
Collapse
Affiliation(s)
- Urbain Weyemi
- Laboratory of Molecular Pharmacology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
32
|
Boudreau HE, Casterline BW, Rada B, Korzeniowska A, Leto TL. Nox4 involvement in TGF-beta and SMAD3-driven induction of the epithelial-to-mesenchymal transition and migration of breast epithelial cells. Free Radic Biol Med 2012; 53:1489-99. [PMID: 22728268 PMCID: PMC3448829 DOI: 10.1016/j.freeradbiomed.2012.06.016] [Citation(s) in RCA: 163] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 12/11/2022]
Abstract
The epithelial-to-mesenchymal transition (EMT) is the development of increased cell plasticity that occurs normally during wound healing and embryonic development and can be coopted for cancer invasion and metastasis. TGF-beta induces EMT but the mechanism is unclear. Our studies suggest that Nox4, a member of the NADPH oxidase (Nox) family, is a source of reactive oxygen species (ROS) affecting cell migration and fibronectin expression, an EMT marker, in normal and metastatic breast epithelial cells. We found that TGF-beta induces Nox4 expression (mRNA and protein) and ROS generation in normal (MCF10A) and metastatic (MDA-MB-231) human breast epithelial cells. Conversely, cells expressing a dominant-negative form of Nox4 or Nox4-targeted shRNA showed significantly lower ROS production on TGF-beta treatment. Expression of a constitutively active TGF-beta receptor type I significantly increased Nox4 promoter activity, mRNA and protein expression, and ROS generation. Nox4 transcriptional regulation by TGF-beta was SMAD3 dependent based on the effect of constitutively active SMAD3 increasing Nox4 promoter activity, whereas dominant-negative SMAD3 or SIS3, a SMAD3-specific inhibitor, had the opposite effect. Furthermore, Nox4 knockdown, dominant-negative Nox4 or SMAD3, or SIS3 blunted TGF-beta induced wound healing and cell migration, whereas cell proliferation was not affected. Our experiments further indicate that Nox4 plays a role in TGF-beta regulation of fibronectin mRNA expression, based on the effects of dominant-negative Nox4 in reducing fibronectin mRNA in TGF-beta-treated MDA-MB-231and MCF10A cells. Collectively, these data indicate that Nox4 contributes to NADPH oxidase-dependent ROS production that may be critical for the progression of the EMT in breast epithelial cells, and thereby has therapeutic implications.
Collapse
Affiliation(s)
- Howard E. Boudreau
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Benjamin W. Casterline
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Balazs Rada
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Agnieszka Korzeniowska
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Thomas L. Leto
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
33
|
Petty WJ, Aklilu M, Varela VA, Lovato J, Savage PD, Miller AA. Reverse translation of phase I biomarker findings links the activity of angiotensin-(1-7) to repression of hypoxia inducible factor-1α in vascular sarcomas. BMC Cancer 2012; 12:404. [PMID: 22963500 PMCID: PMC3495013 DOI: 10.1186/1471-2407-12-404] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 09/03/2012] [Indexed: 12/25/2022] Open
Abstract
Background In a phase I study of angiotensin-(1–7) [Ang-(1–7)], clinical benefit was associated with reduction in plasma placental growth factor (PlGF) concentrations. The current study examines Ang-(1–7) induced changes in biomarkers according to cancer type and investigates mechanisms of action engaged in vitro. Methods Plasma biomarkers were measured prior to Ang-(1–7) administration as well as 1, 2, 3, 4, and 6 hours after treatment. Tests for interaction were performed to determine the impact of cancer type on angiogenic hormone levels. If a positive interaction was detected, treatment-induced biomarker changes for individual cancer types were assessed. To investigate mechanisms of action, in vitro growth assays were performed using a murine endothelioma cell line (EOMA). PCR arrays were performed to identify and statistically validate genes that were altered by Ang-(1–7) treatment in these cells. Results Tests for interaction controlled for dose cohort and clinical response indicated a significant impact of cancer type on post-treatment VEGF and PlGF levels. Following treatment, PlGF levels decreased over time in patients with sarcoma (P = .007). Treatment of EOMA cells with increasing doses of Ang-(1–7) led to significant growth suppression at doses as low as 100 nM. PCR arrays identified 18 genes that appeared to have altered expression after Ang-(1–7) treatment. Replicate analyses confirmed significant changes in 8 genes including reduction in PlGF (P = .04) and hypoxia inducible factor 1α (HIF-1α) expression (P < .001). Conclusions Ang-(1–7) has clinical and pre-clinical activity for vascular sarcomas that is linked to reduced HIF-1α and PlGF expression.
Collapse
Affiliation(s)
- W Jeffrey Petty
- Department of Medicine, Section on Hematology and Oncology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Angiotensin II-induced mitochondrial Nox4 is a major endogenous source of oxidative stress in kidney tubular cells. PLoS One 2012; 7:e39739. [PMID: 22808054 PMCID: PMC3392275 DOI: 10.1371/journal.pone.0039739] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 05/25/2012] [Indexed: 02/07/2023] Open
Abstract
Angiotensin II (Ang II)-induced activation of nicotinamide adenine dinucleotide phosphate (NAD(P)H) oxidase leads to increased production of reactive oxygen species (ROS), an important intracellular second messenger in renal disease. Recent findings suggest that Ang II induces mitochondrial depolarization and further amplifies mitochondrial generation of ROS. We examined the hypothesis that ROS injury mediated by Ang II-induced mitochondrial Nox4 plays a pivotal role in mitochondrial dysfunction in tubular cells and is related to cell survival. In addition, we assessed whether angiotensin (1-7) peptide (Ang-(1-7)) was able to counteract Ang II-induced ROS-mediated cellular injury. Cultured NRK-52E cells were stimulated with 10−6 M Ang II for 24 h with or without Ang-(1-7) or apocynin. Ang II simulated mitochondrial Nox4 and resulted in the abrupt production of mitochondrial superoxide (O2−) and hydrogen peroxide (H2O2). Ang II also induced depolarization of the mitochondrial membrane potential, and cytosolic secretion of cytochrome C and apoptosis-inducing factor (AIF). Ang-(1-7) attenuated Ang II-induced mitochondrial Nox4 expression and apoptosis, and its effect was comparable to that of the NAD(P)H oxidase inhibitor. These findings suggest that Ang II-induced activation of mitochondrial Nox4 is an important endogenous source of ROS, and is related to cell survival. The ACE2-Ang-(1-7)-Mas receptor axis should be investigated further as a novel target of Ang II-mediated ROS injury.
Collapse
|
35
|
Choi DH, Cristóvão AC, Guhathakurta S, Lee J, Joh TH, Beal MF, Kim YS. NADPH oxidase 1-mediated oxidative stress leads to dopamine neuron death in Parkinson's disease. Antioxid Redox Signal 2012; 16:1033-45. [PMID: 22098189 PMCID: PMC3315177 DOI: 10.1089/ars.2011.3960] [Citation(s) in RCA: 137] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
AIM Oxidative stress has long been considered as a major contributing factor in the pathogenesis of Parkinson's disease. However, molecular sources for reactive oxygen species in Parkinson's disease have not been clearly elucidated. Herein, we sought to investigate whether a superoxide-producing NADPH oxidases (NOXs) are implicated in oxidative stress-mediated dopaminergic neuronal degeneration. RESULTS Expression of various Nox isoforms and cytoplasmic components were investigated in N27, rat dopaminergic cells. While most of Nox isoforms were constitutively expressed, Nox1 expression was significantly increased after treatment with 6-hydroxydopamine. Rac1, a key regulator in the Nox1 system, was also activated. Striatal injection of 6-hydroxydopamine increased Nox1 expression in dopaminergic neurons in the rat substantia nigra. Interestingly, it was localized into the nucleus, and immunostaining for DNA oxidative stress marker, 8-oxo-dG, was increased. Nox1 expression was also found in the nucleus of dopaminergic neurons in the substantia nigra of Parkinson's disease patients. Adeno-associated virus-mediated Nox1 knockdown or Rac1 inhibition reduced 6-hydroxydopamine-induced oxidative DNA damage and dopaminergic neuronal degeneration significantly. INNOVATION Nox1/Rac1 could serve as a potential therapeutic target for Parkinson's disease. CONCLUSION We provide evidence that dopaminergic neurons are equipped with the Nox1/Rac1 superoxide-generating system. Stress-induced Nox1/Rac1 activation causes oxidative DNA damage and neurodegeneration. Reduced dopaminergic neuronal death achieved by targeting Nox1/Rac1, emphasizes the impact of oxidative stress caused by this system on the pathogenesis and therapy in Parkinson's disease.
Collapse
Affiliation(s)
- Dong-Hee Choi
- Neurology/Neuroscience Department, Weill Medical College of Cornell University, New York, New York, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Song P, Zou MH. Regulation of NAD(P)H oxidases by AMPK in cardiovascular systems. Free Radic Biol Med 2012; 52:1607-19. [PMID: 22357101 PMCID: PMC3341493 DOI: 10.1016/j.freeradbiomed.2012.01.025] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Revised: 01/25/2012] [Accepted: 01/27/2012] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are ubiquitously produced in cardiovascular systems. Under physiological conditions, ROS/RNS function as signaling molecules that are essential in maintaining cardiovascular function. Aberrant concentrations of ROS/RNS have been demonstrated in cardiovascular diseases owing to increased production or decreased scavenging, which have been considered common pathways for the initiation and progression of cardiovascular diseases such as atherosclerosis, hypertension, (re)stenosis, and congestive heart failure. NAD(P)H oxidases are primary sources of ROS and can be induced or activated by all known cardiovascular risk factors. Stresses, hormones, vasoactive agents, and cytokines via different signaling cascades control the expression and activity of these enzymes and of their regulatory subunits. But the molecular mechanisms by which NAD(P)H oxidase is regulated in cardiovascular systems remain poorly characterized. Investigations by us and others suggest that adenosine monophosphate-activated protein kinase (AMPK), as an energy sensor and modulator, is highly sensitive to ROS/RNS. We have also obtained convincing evidence that AMPK is a physiological suppressor of NAD(P)H oxidase in multiple cardiovascular cell systems. In this review, we summarize our current understanding of how AMPK functions as a physiological repressor of NAD(P)H oxidase.
Collapse
Affiliation(s)
| | - Ming-Hui Zou
- To whom correspondence should be addressed: Ming-Hui Zou, M.D., Ph.D., Department of Medicine, University of Oklahoma Health Science Center, 941 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA, Phone: 405-271-3974, Fax: 405-271-3973,
| |
Collapse
|
37
|
Choi J. Oxidative stress, endogenous antioxidants, alcohol, and hepatitis C: pathogenic interactions and therapeutic considerations. Free Radic Biol Med 2012; 52:1135-50. [PMID: 22306508 DOI: 10.1016/j.freeradbiomed.2012.01.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 01/04/2012] [Accepted: 01/12/2012] [Indexed: 12/16/2022]
Abstract
Hepatitis C virus (HCV) is a blood-borne pathogen that was identified as an etiologic agent of non-A, non-B hepatitis in 1989. HCV is estimated to have infected at least 170 million people worldwide. The majority of patients infected with HCV do not clear the virus and become chronically infected, and chronic HCV infection increases the risk for hepatic steatosis, cirrhosis, and hepatocellular carcinoma. HCV induces oxidative/nitrosative stress from multiple sources, including inducible nitric oxide synthase, the mitochondrial electron transport chain, hepatocyte NAD(P)H oxidases, and inflammation, while decreasing glutathione. The cumulative oxidative burden is likely to promote both hepatic and extrahepatic conditions precipitated by HCV through a combination of local and more distal effects of reactive species, and clinical, animal, and in vitro studies strongly point to a role of oxidative/nitrosative stress in HCV-induced pathogenesis. Oxidative stress and hepatopathogenesis induced by HCV are exacerbated by even low doses of alcohol. Alcohol and reactive species may have other effects on hepatitis C patients such as modulation of the host immune system, viral replication, and positive selection of HCV sequence variants that contribute to antiviral resistance. This review summarizes the current understanding of redox interactions of HCV, outlining key experimental findings, directions for future research, and potential applications to therapy.
Collapse
Affiliation(s)
- Jinah Choi
- Department of Molecular Cell Biology, School of Natural Sciences, University of California at Merced, Merced, CA 95343, USA.
| |
Collapse
|
38
|
Auchinvole CAR, Richardson P, McGuinnes C, Mallikarjun V, Donaldson K, McNab H, Campbell CJ. Monitoring intracellular redox potential changes using SERS nanosensors. ACS NANO 2012; 6:888-896. [PMID: 22165857 DOI: 10.1021/nn204397q] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Redox homeostasis and signaling are critically important in the regulation of cell function. There are significant challenges in quantitatively measuring intracellular redox potentials, and in this paper, we introduce a new approach. Our approach is based on the use of nanosensors which comprise molecules that sense the local redox potential, assembled on a gold nanoshell. Since the Raman spectrum of the sensor molecule changes depending on its oxidation state and since the nanoshell allows a huge enhancement of the Raman spectrum, intracellular potential can be calculated by a simple optical measurement. The nanosensors can be controllably delivered to the cytoplasm, without any toxic effects, allowing redox potential to be monitored in a reversible, non-invasive manner over a previously unattainable potential range encompassing both superphysiological and physiological oxidative stress.
Collapse
Affiliation(s)
- Craig A R Auchinvole
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3JJ, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
39
|
Drummond GR, Selemidis S, Griendling KK, Sobey CG. Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nat Rev Drug Discov 2011; 10:453-71. [PMID: 21629295 PMCID: PMC3361719 DOI: 10.1038/nrd3403] [Citation(s) in RCA: 690] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NADPH oxidases are a family of enzymes that generate reactive oxygen species (ROS). The NOX1 (NADPH oxidase 1) and NOX2 oxidases are the major sources of ROS in the artery wall in conditions such as hypertension, hypercholesterolaemia, diabetes and ageing, and so they are important contributors to the oxidative stress, endothelial dysfunction and vascular inflammation that underlies arterial remodelling and atherogenesis. In this Review, we advance the concept that compared to the use of conventional antioxidants, inhibiting NOX1 and NOX2 oxidases is a superior approach for combating oxidative stress. We briefly describe some common and emerging putative NADPH oxidase inhibitors. In addition, we highlight the crucial role of the NADPH oxidase regulatory subunit, p47phox, in the activity of vascular NOX1 and NOX2 oxidases, and suggest how a better understanding of its specific molecular interactions may enable the development of novel isoform-selective drugs to prevent or treat cardiovascular diseases.
Collapse
Affiliation(s)
- Grant R Drummond
- Vascular Biology & Immunopharmacology Group, Department of Pharmacology, Monash University, Victoria 3800, Australia.
| | | | | | | |
Collapse
|
40
|
Spencer NY, Yan Z, Boudreau RL, Zhang Y, Luo M, Li Q, Tian X, Shah AM, Davisson RL, Davidson B, Banfi B, Engelhardt JF. Control of hepatic nuclear superoxide production by glucose 6-phosphate dehydrogenase and NADPH oxidase-4. J Biol Chem 2011; 286:8977-87. [PMID: 21212270 DOI: 10.1074/jbc.m110.193821] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Redox-regulated signal transduction is coordinated by spatially controlled production of reactive oxygen species within subcellular compartments. The nucleus has long been known to produce superoxide (O(2)(·-)); however, the mechanisms that control this function remain largely unknown. We have characterized molecular features of a nuclear superoxide-producing system in the mouse liver. Using electron paramagnetic resonance, we investigated whether several NADPH oxidases (NOX1, 2, and 4) and known activators of NOX (Rac1, Rac2, p22(phox), and p47(phox)) contribute to nuclear O(2)(·-) production in isolated hepatic nuclei. Our findings demonstrate that NOX4 most significantly contributes to hepatic nuclear O(2)(·-) production that utilizes NADPH as an electron donor. Although NOX4 protein immunolocalized to both nuclear membranes and intranuclear inclusions, fluorescent detection of NADPH-dependent nuclear O(2)(·-) predominantly localized to the perinuclear space. Interestingly, NADP(+) and G6P also induced nuclear O(2)(·-) production, suggesting that intranuclear glucose-6-phosphate dehydrogenase (G6PD) can control NOX4 activity through nuclear NADPH production. Using G6PD mutant mice and G6PD shRNA, we confirmed that reductions in nuclear G6PD enzyme decrease the ability of hepatic nuclei to generate O(2)(·-) in response to NADP(+) and G6P. NOX4 and G6PD protein were also observed in overlapping microdomains within the nucleus. These findings provide new insights on the metabolic pathways for substrate regulation of nuclear O(2)(·-) production by NOX4.
Collapse
Affiliation(s)
- Netanya Y Spencer
- Department of Anatomy and Cell Biology, College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The Noxes (NADPH oxidases) are a family of ROS (reactive oxygen species)-generating enzymes. Of the seven family members, four have been identified as important sources of ROS in the vasculature: Nox1, Nox2, Nox4 and Nox5. Although Nox isoforms can be influenced by the same stimulus and co-localize in cellular compartments, their tissue distribution, subcellular regulation, requirement for cofactors and NADPH oxidase subunits and ability to generate specific ROS differ, which may help to understand the multiplicity of biological functions of these oxidases. Nox4 and Nox5 are the newest isoforms identified in the vasculature. Nox4 is the major isoform expressed in renal cells and appear to produce primarily H2O2. The Nox5 isoform produces ROS in response to increased levels of intracellular Ca2+ and does not require the other NADPH oxidase subunits for its activation. The present review focuses on these unique Noxes, Nox4 and Nox5, and provides novel concepts related to the regulation and interaction in the vasculature, and discusses new potential roles for these isoforms in vascular biology.
Collapse
|
42
|
Cho SO, Lim JW, Kim KH, Kim H. Diphenyleneiodonium inhibits the activation of mitogen-activated protein kinases and the expression of monocyte chemoattractant protein-1 in Helicobacter pylori-infected gastric epithelial AGS cells. Inflamm Res 2010; 60:501-7. [PMID: 21181544 DOI: 10.1007/s00011-010-0297-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 11/13/2010] [Accepted: 12/02/2010] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE To investigate whether NADPH oxidase induces MCP-1 expression and the activation of mitogen-activated protein kinases (MAPKs) in H. pylori-infected gastric epithelial cells. MATERIAL H. pylori in Korean isolates, human gastric epithelial AGS cells TREATMENT AGS cells pretreated with or without an NADPH oxidase inhibitor diphenyleneiodonium (DPI) are cultured in the presence of H. pylori at a bacterium/cell ratio of 300:1. METHODS Reactive oxygen species (ROS) and MCP-1 were determined by confocal microscopy and enzyme-linked immonosorbent assay. NADPH oxidase activity was measured by lucigenin assay. mRNA expression of MCP-1 was analyzed by reverse transcription-polymerase chain reaction. Levels of MAPKs were assessed by Western blot analysis. RESULTS H. pylori induced increase in ROS, NADPH oxidase activity, MCP-1 expression, and the activation of MAPKs including extracellular signal-regulated kinases, p38, and jun N-terminal kinases in AGS cells, which was inhibited by DPI. CONCLUSION Inhibiting NADPH oxidase by DPI suppresses H. pylori-induced activation of MAPKs and MCP-1 expression in AGS cells.
Collapse
Affiliation(s)
- Soon Ok Cho
- Department of Pharmacology, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, 120-752, Korea
| | | | | | | |
Collapse
|
43
|
Liu RM, Choi J, Wu JH, Gaston Pravia KA, Lewis KM, Brand JD, Mochel NSR, Krzywanski DM, Lambeth JD, Hagood JS, Forman HJ, Thannickal VJ, Postlethwait EM. Oxidative modification of nuclear mitogen-activated protein kinase phosphatase 1 is involved in transforming growth factor beta1-induced expression of plasminogen activator inhibitor 1 in fibroblasts. J Biol Chem 2010; 285:16239-47. [PMID: 20228065 DOI: 10.1074/jbc.m110.111732] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor beta (TGF-beta) stimulates reactive oxygen species (ROS) production in various cell types, which mediates many of the effects of TGF-beta. The molecular mechanisms whereby TGF-beta increases ROS production and ROS modulate the signaling processes of TGF-beta, however, remain poorly defined. In this study, we show that TGF-beta1 stimulates NADPH oxidase 4 (Nox4) expression and ROS generation in the nucleus of murine embryo fibroblasts (NIH3T3 cells). This is associated with an increase in protein thiol modification and inactivation of MAPK phosphatase 1 (MKP-1), a nuclear phosphatase. Furthermore, knockdown of MKP-1 using small interfering RNA enhances TGF-beta1-induced phosphorylation of JNK and p38 as well as the expression of plasminogen activator inhibitor 1 (PAI-1), a TGF-beta-responsive gene involved in the pathogenesis of many diseases. Knockdown of Nox4 with Nox4 small interfering RNA, on the other hand, reduces TGF-beta1-stimulated ROS production, p38 phosphorylation, and PAI-1 expression. TGF-beta also increased the nuclear level of Nox4 protein as well as PAI-1 expression in human lung fibroblasts (CCL-210 cells), suggesting that TGF-beta may induce PAI-1 expression by a similar mechanism in human lung fibroblasts. In summary, in this study we have identified nuclear MAPK phosphatase MKP-1 as a novel molecular target of ROS in TGF-beta signaling pathways. Our data suggest that increased generation of ROS by Nox4 mediates TGF-beta1-induced PAI-1 gene expression at least in part through oxidative modification and inhibition of MKP-1 leading to a sustained activation of JNK and p38 MAPKs.
Collapse
Affiliation(s)
- Rui-Ming Liu
- Department of Environmental Health Sciences, School of Public Health, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|