51
|
Tejero J, Shiva S, Gladwin MT. Sources of Vascular Nitric Oxide and Reactive Oxygen Species and Their Regulation. Physiol Rev 2019; 99:311-379. [PMID: 30379623 PMCID: PMC6442925 DOI: 10.1152/physrev.00036.2017] [Citation(s) in RCA: 337] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/30/2018] [Accepted: 05/06/2018] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) is a small free radical with critical signaling roles in physiology and pathophysiology. The generation of sufficient NO levels to regulate the resistance of the blood vessels and hence the maintenance of adequate blood flow is critical to the healthy performance of the vasculature. A novel paradigm indicates that classical NO synthesis by dedicated NO synthases is supplemented by nitrite reduction pathways under hypoxia. At the same time, reactive oxygen species (ROS), which include superoxide and hydrogen peroxide, are produced in the vascular system for signaling purposes, as effectors of the immune response, or as byproducts of cellular metabolism. NO and ROS can be generated by distinct enzymes or by the same enzyme through alternate reduction and oxidation processes. The latter oxidoreductase systems include NO synthases, molybdopterin enzymes, and hemoglobins, which can form superoxide by reduction of molecular oxygen or NO by reduction of inorganic nitrite. Enzymatic uncoupling, changes in oxygen tension, and the concentration of coenzymes and reductants can modulate the NO/ROS production from these oxidoreductases and determine the redox balance in health and disease. The dysregulation of the mechanisms involved in the generation of NO and ROS is an important cause of cardiovascular disease and target for therapy. In this review we will present the biology of NO and ROS in the cardiovascular system, with special emphasis on their routes of formation and regulation, as well as the therapeutic challenges and opportunities for the management of NO and ROS in cardiovascular disease.
Collapse
Affiliation(s)
- Jesús Tejero
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Sruti Shiva
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark T Gladwin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh , Pittsburgh, Pennsylvania ; Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania ; Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania ; and Department of Medicine, Center for Metabolism and Mitochondrial Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
52
|
Ferreira CA, Ni D, Rosenkrans ZT, Cai W. Scavenging of reactive oxygen and nitrogen species with nanomaterials. NANO RESEARCH 2018; 11:4955-4984. [PMID: 30450165 PMCID: PMC6233906 DOI: 10.1007/s12274-018-2092-y] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/01/2018] [Accepted: 05/04/2018] [Indexed: 05/03/2023]
Abstract
Reactive oxygen and nitrogen species (RONS) are essential for normal physiological processes and play important roles in cell signaling, immunity, and tissue homeostasis. However, excess radical species are implicated in the development and augmented pathogenesis of various diseases. Several antioxidants may restore the chemical balance, but their use is limited by disappointing results of clinical trials. Nanoparticles are an attractive therapeutic alternative because they can change the biodistribution profile of antioxidants, and possess intrinsic ability to scavenge RONS. Herein, we review the types of RONS, how they are implicated in several diseases, and the types of nanoparticles with inherent antioxidant capability, their mechanisms of action, and their biological applications.
Collapse
Affiliation(s)
- Carolina A. Ferreira
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dalong Ni
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Weibo Cai
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
53
|
Du S, Miao J, Zhu Z, Xu E, Shi L, Ai S, Wang F, Kang X, Chen H, Lu X, Guan W, Xia X. NADPH oxidase 4 regulates anoikis resistance of gastric cancer cells through the generation of reactive oxygen species and the induction of EGFR. Cell Death Dis 2018; 9:948. [PMID: 30237423 PMCID: PMC6148243 DOI: 10.1038/s41419-018-0953-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 07/22/2018] [Accepted: 08/03/2018] [Indexed: 01/17/2023]
Abstract
Anoikis is a type of programmed cell death induced by detachment from the extracellular matrix. In cancer cells, anoikis resistance is essential for cancer cell survival in blood circulation and distant metastasis. However, the mechanisms behind anoikis resistance of gastric cancer remain largely unknown. Herein, we demonstrate that NADPH oxidase 4 (NOX4) expression and reactive oxygen species (ROS) generation are upregulated in suspension gastric cell cultures compared with adherent cultures. Silencing of NOX4 decreases ROS generation and downregulates EGFR, sensitizing cells to anoikis. NOX4 overexpression upregulates ROS and EGFR levels and promotes anoikis resistance. NOX4 depletion inhibits gastric cancer survival in blood circulation and attenuates distant metastasis. NOX4 expression is correlated with EGFR expression in patients. In conclusion, induction of NOX4 expression by detachment promotes anoikis resistance of gastric cancer through ROS generation and downstream upregulation of EGFR, which is critical for the metastatic progression of gastric cancer.
Collapse
Affiliation(s)
- Shangce Du
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China.,Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - Ji Miao
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China.,Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - Zhouting Zhu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - En Xu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - Linsen Shi
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - Shichao Ai
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - Feng Wang
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China.,Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - Xing Kang
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China.,Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - Hong Chen
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China.,Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - Xiaofeng Lu
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China.,Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China
| | - Wenxian Guan
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China. .,Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China.
| | - Xuefeng Xia
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China. .,Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, 210008, Nanjing, Jiangsu, P. R. China.
| |
Collapse
|
54
|
Drevet S, Gavazzi G, Grange L, Dupuy C, Lardy B. Reactive oxygen species and NADPH oxidase 4 involvement in osteoarthritis. Exp Gerontol 2018; 111:107-117. [PMID: 30012342 DOI: 10.1016/j.exger.2018.07.007] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 01/04/2023]
Abstract
Osteoarthritis (OA) is a degenerative chronic disease affecting >300,000 million people around the world as of 2016. Symptomatic measures exist, but there are hardly any curative treatments available. Disruption of the cartilage homeostasis in favor of catabolism leads to cartilage destruction. ROS-macromolecular-induced damage is significantly greater in OA cartilage and OA is described as low-grade chronic systemic inflammation. This review aimed to assess the critical role of cartilage ageing and oxidative stress in the OA process, focusing in particular on NADPH oxidase and especially Nox4 involvement. With age, hypertrophic senescent cells with an altered redox cell profile accumulated. Chondrocytes are more sensitive to oxidant-mediators and the serum level of pro-inflammatory mediators increases. Age-related advanced glycation end products impact on extra cellular matrix (ECM) properties leading to the apoptosis of chondrocytes. A focus on NADPH oxidase-mediated-ROS signaling highlighted the very specific Nox4 isoform, which plays a role on the final common pathway targeting chondrocyte cells. IL-1β-mediated Nox4 stimulation induced an increase in the levels released by the chondrocyte of MMP-1 and MMP-13 proteins, which are involved in ECM degradation. In comparison with the other Nox isoforms, Nox4 remains unusual, since it is constitutively active, does not depend on cytosolic activator proteins and seems to generate H2O2 thanks to the specific conformation of the Nox4 E-loop. Nox4-induced ROS production appears an essential actor in the OA process and it could be relevant to focus on this target in the aim of discovering and developing new therapeutic strategies.
Collapse
Affiliation(s)
- S Drevet
- Grenoble Alpes University Hospital, Orthogeriatric Unit, Geriatric Department, Grenoble Alpes University, GREPI UGA-EFS EA7408, Boulevard de la Chantourne, 38043 Grenoble Cedex 1, France.
| | - G Gavazzi
- Grenoble Alpes University Hospital, Acute Geriatric Medicine Unit, Geriatric Department, Grenoble Alpes University, GREPI UGA-EFS EA7408, Boulevard de la Chantourne, 38043 Grenoble Cedex 1, France.
| | - L Grange
- Grenoble Alpes University Hospital, Rheumatology Department, Hopital Sud, GREPI UGA-EFS EA7408, 19 avenue de Kimberley, 38130 Echirolles, France.
| | - C Dupuy
- Institut Gustave Roussy, UMR 8200 CNRS "Stabilité génétique et Oncogenèse", 114 rue Edouard Vaillant, 94805 Villejuif Cedex, France.
| | - B Lardy
- Grenoble Alpes University Hospital, Biology Department, Grenoble Alpes University, GREPI UGA-EFS EA7408, Boulevard de la Chantourne, 38043 Grenoble Cedex 1, France.
| |
Collapse
|
55
|
Zandalinas SI, Mittler R. ROS-induced ROS release in plant and animal cells. Free Radic Biol Med 2018; 122:21-27. [PMID: 29203327 DOI: 10.1016/j.freeradbiomed.2017.11.028] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/27/2017] [Accepted: 11/30/2017] [Indexed: 01/08/2023]
Abstract
Reactive oxygen species (ROS) play a key signaling role in plant and animal cells. Among the many cellular mechanisms used to generate and transduce ROS signals, ROS-induced ROS release (RIRR) is emerging as an important pathway involved in different human pathologies and plant responses to environmental stress. RIRR is a process in which one cellular compartment or organelle generates or releases ROS, triggering the enhanced production or release of ROS by another compartment or organelle. It was initially described in animal cells and proposed to mediate mitochondria-to-mitochondria communication, but later expanded to include communication between mitochondria and plasma membrane-localized NADPH oxidases. In plants a process of RIRR was demonstrated to mediate long distance rapid systemic signaling in response to biotic and abiotic stress. This process is thought to involve the enhanced production of ROS by one cell that triggers the enhanced production of ROS by a neighboring cell in a process that propagates the enhanced "ROS production state" all the way from one part of the plant to another. In contrast to the intracellular nature of the RIRR process of animal cells, the plant RIRR process is therefore primarily studied at the cell-to-cell communication level. Studies on intracellular (organelle-to-organelle, or organelle-to-NADPH oxidase) RIRR pathways are very scarce in plants, whereas studies on cell-to-cell RIRR are very scarce in animals. Here we will attempt to highlight what is known in both systems and what each system can learn from the other.
Collapse
Affiliation(s)
- Sara I Zandalinas
- Department of Biological Sciences, College of Arts and Sciences, University of North Texas, 1155 Union Circle #305220, Denton, TX 76203-5017, USA
| | - Ron Mittler
- Department of Biological Sciences, College of Arts and Sciences, University of North Texas, 1155 Union Circle #305220, Denton, TX 76203-5017, USA.
| |
Collapse
|
56
|
Chen F, Wei G, Xu J, Ma X, Wang Q. Naringin ameliorates the high glucose-induced rat mesangial cell inflammatory reaction by modulating the NLRP3 Inflammasome. Altern Ther Health Med 2018; 18:192. [PMID: 29929501 PMCID: PMC6014005 DOI: 10.1186/s12906-018-2257-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/13/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND The Nucleotide binding and oligomerization domain-like receptorfamily pyrin domain-containing 3 (NLRP3)-inflammasome plays an important role in various diseases, including a variety of kidney diseases. Naringin exhibits anti-inflammatory and anti-oxidation effects among others, but its specific mechanisms are not clear. We investigated the expression of the NLRP3-inflammasome under high-glucose conditions, assessed the effects of naringin on that process, and further elucidated the role of naringin in the pathogenesis of diabetic kidney disease(DKD). METHODS To assess the therapeutic potential of naringin and the mechanisms involved, we cultured rat glomerular mesangial cells and grouped them according to different glucose concentrations, different action times, different concentrations of MCC950, and different concentrations of naringin.The cell proliferation was measured by MTT assay. The expression of Interleukin-1β(IL-1β) and Interleukin18 (IL-18) in the cell supernatant were detected by ELISA. The expression and activity of NLPR3, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and Caspase-1 were detected by Western Blot. RESULTS The expressions of NLRP3, ASC, caspase-1, IL-1β, and IL-18 in rat glomerular mesangial cells were significantly higher in the high glucose (HG) group than in the control normal glucose (NG) group and exhibited time-dependence activity. The expression levels of NLRP3, caspase-1, IL-1β, and IL-18 in different treatment groups were significantly lower compared with the HG group after 48 h of MCC950 pre-treatment (p < 0.05). Pre-treatment with naringin produced the same results. Naringin also inhibited the proliferation of cells. CONCLUSIONS The NLRP3-inflammasome potentially plays a role in the process of activation and inflammation of glomerular mesangial cells as induced by high-glucose conditions. Naringin inhibited the proliferation of cells that were induced by high glucose. Further, it reduced the expression of inflammatory factors that are mediated by NLRP3 through the NLRP3-caspase-1-IL-1β/IL-18 signaling pathway, which makes naringin a potentially novel treatment for DKD disease.
Collapse
|
57
|
Shih WL, Chang CD, Chen HT, Fan KK. Antioxidant activity and leukemia initiation prevention in vitro and in vivo by N-acetyl-L-cysteine. Oncol Lett 2018; 16:2046-2052. [PMID: 30008899 DOI: 10.3892/ol.2018.8864] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/22/2018] [Indexed: 01/12/2023] Open
Abstract
N-acetyl-L-cysteine (NAC) is the most abundant water-soluble component of garlic. No study to date has studied the leukemia prevention ability of NAC in mouse systemic leukemia model. The current study aimed to investigate the leukemia initiation prevention potential of NAC in a mouse model. The cytotoxic concentration of NAC was determined first in HL-60 cells, and its in vivo activity was studied in a mouse acute myelocytic leukemia model with WEHI-3 leukemia cells. The results showed that a non-toxic concentration of NAC efficiently scavenged free-radicals, lowered lipid peroxidation and reduced DNA damage induced by hydrogen peroxide in a cultured HL-60 leukemia cell line. NAC also elevated the cellular antioxidant enzyme activity significantly. Furthermore, NAC prevented mouse death induced by injection of murine WEHI-3 leukemia cells and reduced organ damage, as well as activated antioxidant mechanisms. The results of this study provided strong evidence that NAC may have potential benefits in terms of elevating antioxidant activity and preventing leukemia initiation.
Collapse
Affiliation(s)
- Wen-Ling Shih
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan, R.O.C
| | - Ching-Dong Chang
- Department of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan, R.O.C
| | - Hsiu-Tsu Chen
- Department of Fashion Design and Management, National Pingtung University of Science and Technology, Pingtung 91201, Taiwan, R.O.C.,Graduate School of Design, National Yunlin University of Science and Technology, Yunlin 64002, Taiwan, R.O.C
| | - Kuo-Kuang Fan
- Graduate School of Design, National Yunlin University of Science and Technology, Yunlin 64002, Taiwan, R.O.C
| |
Collapse
|
58
|
Abstract
Cancer cell migration is essential for metastasis, during which cancer cells move through the tumor and reach the blood vessels. In vivo, cancer cells are exposed to contact guidance and chemotactic cues. Depending on the strength of such cues, cells will migrate in a random or directed manner. While similar cues may also stimulate cell proliferation, it is not clear whether cell cycle progression affects migration of cancer cells and whether this effect is different in random versus directed migration. In this study, we tested the effect of cell cycle progression on contact guided migration in 2D and 3D environments, in the breast carcinoma cell line, FUCCI-MDA-MB-231. The results were quantified from live cell microscopy images using the open source lineage editing and validation image analysis tools (LEVER). In 2D, cells were placed inside 10 μm-wide microchannels to stimulate contact guidance, with or without an additional chemotactic gradient of the soluble epidermal growth factor. In 3D, contact guidance was modeled by aligned collagen fibers. In both 2D and 3D, contact guidance was cell cycle-dependent, while the addition of the chemo-attractant gradient in 2D increased cell velocity and persistence in directionally migrating cells, regardless of their cell cycle phases. In both 2D and 3D contact guidance, cells in the G1 phase of the cell cycle outperformed cells in the S/G2 phase in terms of migration persistence and instantaneous velocity. These data suggest that in the presence of contact guidance cues in vivo, breast carcinoma cells in the G1 phase of the cell cycle may be more efficient in reaching the neighboring vasculature.
Collapse
Affiliation(s)
| | - Edgar Cardenas De La Hoz
- Department of Electrical and Computer Engineering, College of Engineering, Drexel University, Philadelphia, Pennsylvania 19104, USA
| | - Andrew R Cohen
- Department of Electrical and Computer Engineering, College of Engineering, Drexel University, Philadelphia, Pennsylvania 19104, USA
| | - Bojana Gligorijevic
- Bioengineering department, College of Engineering, Temple University, Philadelphia, Pennsylvania 19122, USA.,Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| |
Collapse
|
59
|
Shashni B, Nagasaki Y. Nitroxide radical-containing nanoparticles attenuate tumorigenic potential of triple negative breast cancer. Biomaterials 2018; 178:48-62. [PMID: 29908344 DOI: 10.1016/j.biomaterials.2018.05.042] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/26/2018] [Accepted: 05/25/2018] [Indexed: 02/07/2023]
Abstract
The critical importance of reactive oxygen species (ROS) as oncogene activators and essential secondary messengers in cancer cell survival have been widely reported. Since oxidative stress has been implicated as being pivotal in various cancers, antioxidant therapy seems an apt strategy to abrogate ROS-mediated cellular processes to attenuate cancers. We therefore synthesized ROS scavenging nitroxide radical-containing nanoparticles (RNPs); pH insensitive RNPO and pH sensitive RNPN, to impede the proliferative and metastatic characteristics of the triple negative breast cancer cell line, MDA-MB-231, both in vitro and in vivo. RNPs significantly curtailed the proliferative and clonogenic potential of MDA-MB-231 and MCF-7 cell lines. Inhibition of ROS-mediated migratory and invasive characteristics of MDA-MB-231, via down regulation of NF-κB and MMP-2, was also confirmed. Furthermore, a significant anti-tumor and anti-metastatic potential of RNPs was observed in an MDA-MB-231 mouse xenograft model. Such tumoricidal effects of RNPs were attained with negligible adverse effects, compared to conventional low molecular weight antioxidants, TEMPOL. Thus, the tumoricidal effects of RNPs are suggestive of insights on precedence of nanoparticle-based therapeutics over current low molecular weight antioxidants to curtail ROS-induced tumorigenesis of various cancers.
Collapse
Affiliation(s)
- Babita Shashni
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan
| | - Yukio Nagasaki
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan; Master's School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan; Center for Research in Isotope and Environmental Dynamics (CRiED), University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8573, Japan.
| |
Collapse
|
60
|
Genna A, Gil-Henn H. FAK family kinases: The Yin and Yang of cancer cell invasiveness. Mol Cell Oncol 2018; 5:e1449584. [PMID: 30250911 PMCID: PMC6149990 DOI: 10.1080/23723556.2018.1449584] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 03/03/2018] [Accepted: 03/03/2018] [Indexed: 01/14/2023]
Abstract
Proline-rich tyrosine kinase 2 (PYK2, also known as Pyk2) and its closely related focal adhesion kinase (FAK) modulate cancer cell invasion by coordinating the balance between focal adhesion-mediated migration and invadopodia-dependent extracellular matrix invasion. Our recent findings present Pyk2 and FAK as novel mediators of breast cancer invasiveness and as potential targets for blocking breast cancer metastasis.
Collapse
Affiliation(s)
- Alessandro Genna
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
61
|
NEDD9 stimulated MMP9 secretion is required for invadopodia formation in oral squamous cell carcinoma. Oncotarget 2018; 9:25503-25516. [PMID: 29876004 PMCID: PMC5986644 DOI: 10.18632/oncotarget.25347] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 04/24/2018] [Indexed: 12/19/2022] Open
Abstract
Neural precursor cell expressed developmentally downregulated 9 (NEDD9) is a component of the metastatic signatures of melanoma, breast cancer, glioblastoma, lung cancer and head and neck squamous cell carcinoma (HNSCC). Here we tested the efficacy of NEDD9's domains in stimulating matrix metalloproteinase (MMP) secretion and invadopodia formation in cells stably expressing various NEDD9 mutants. Replacement of the 13 YxxP motif substrate domain (SD) tyrosines and the C-terminal Y629 with phenylalanines (F14NEDD9) eliminated tyrosine phosphorylation, MMP9 secretion and loss of invadopodia formation. Mutation of the N-terminal SH3 domain Y12 to glutamic acid (Y12ENEDD9) or phenylalanine (Y12FNEDD9) reduced MMP9 secretion and inhibited invadopodia formation. SH3 domain deletion (∆SH3NEDD9) resulted in the loss of MMP9 secretion and a lack of invadopodia formation. The SH3–SD domain (SSNEDD9) construct exhibited tyrosine phosphorylation and stimulated MMP9 secretion, as did ∆CTNEDD9 which lacked the C-terminus (∆C-terminal; ∆CT). E13NEDD9 expression blocked MMP9 secretion and invadopodia formation. MICAL1 (molecule interacting with Cas-L1) silencing with a short hairpin RNA reduced MMP9 secretion, vimentin and E-cadherin levels while increasing N-cadherin and Rab6 levels, consistent with reduced invasive behavior. These findings indicate that NEDD9 SD phosphorylation and SH3 domain interactions are necessary for increasing MMP9 secretion and invadopodia formation.
Collapse
|
62
|
Meirson T, Gil-Henn H. Targeting invadopodia for blocking breast cancer metastasis. Drug Resist Updat 2018; 39:1-17. [PMID: 30075834 DOI: 10.1016/j.drup.2018.05.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 05/04/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022]
Abstract
Dissemination of cancer cells from the primary tumor and their spread to distant sites of the body is the leading cause of mortality in metastatic cancer patients. Metastatic cancer cells invade surrounding tissues and blood vessels by forming F-actin-rich protrusions known as invadopodia, which degrade the extracellular matrix and enable invasion of tumor cells through it. Invadopodia have now been observed in vivo, and recent evidence demonstrates direct molecular links between assembly of invadopodia and cancer metastasis in both mouse models and in human patients. While significant progress has been achieved in the last decade in understanding the molecular mechanisms and signaling pathways regulating invadopodia formation and function, the application of this knowledge to development of prognostic and therapeutic approaches for cancer metastasis has not been discussed before. Here, we provide a detailed overview of current prognostic markers and tests for cancer metastasis and discuss their advantages, disadvantages, and their predicted efficiency. Using bioinformatic patient database analysis, we demonstrate, for the first time, a significant correlation between invadopodia-associated genes to breast cancer metastasis, suggesting that invadopodia could be used as both a prognostic marker and as a therapeutic target for blocking cancer metastasis. We include here a novel network interaction map of invadopodia-associated proteins with currently available inhibitors, demonstrating a central role for the recently identified EGFR-Pyk2-Src-Arg-cortactin invadopodial pathway, to which re-purposing of existent inhibitors could be used to block breast cancer metastasis. We then present an updated overview of current cancer-related clinical trials, demonstrating the negligible number of trials focusing on cancer metastasis. We also discuss the difficulties and complexity of performing cancer metastasis clinical trials, and the possible development of anti-metastasis drug resistance when using a prolonged preventive treatment with invadopodia inhibitors. This review presents a new perspective on invadopodia-mediated tumor invasiveness and may lead to the development of novel prognostic and therapeutic approaches for cancer metastasis.
Collapse
Affiliation(s)
- Tomer Meirson
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel; Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel.
| |
Collapse
|
63
|
From Oxidative Stress Damage to Pathways, Networks, and Autophagy via MicroRNAs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4968321. [PMID: 29849898 PMCID: PMC5932428 DOI: 10.1155/2018/4968321] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 03/04/2018] [Indexed: 11/24/2022]
Abstract
Oxidative stress can alter the expression level of many microRNAs (miRNAs), but how these changes are integrated and related to oxidative stress responses is poorly understood. In this article, we addressed this question by using in silico tools. We reviewed the literature for miRNAs whose expression is altered upon oxidative stress damage and used them in combination with various databases and software to predict common gene targets of oxidative stress-modulated miRNAs and affected pathways. Furthermore, we identified miRNAs that simultaneously target the predicted oxidative stress-modulated miRNA gene targets. This generated a list of novel candidate miRNAs potentially involved in oxidative stress responses. By literature search and grouping of pathways and cellular responses, we could classify these candidate miRNAs and their targets into a larger scheme related to oxidative stress responses. To further exemplify the potential of our approach in free radical research, we used our explorative tools in combination with ingenuity pathway analysis to successfully identify new candidate miRNAs involved in the ubiquitination process, a master regulator of cellular responses to oxidative stress and proteostasis. Lastly, we demonstrate that our approach may also be useful to identify novel candidate connections between oxidative stress-related miRNAs and autophagy. In summary, our results indicate novel and important aspects with regard to the integrated biological roles of oxidative stress-modulated miRNAs and demonstrate how this type of in silico approach can be useful as a starting point to generate hypotheses and guide further research on the interrelation between miRNA-based gene regulation, oxidative stress signaling pathways, and autophagy.
Collapse
|
64
|
Bartas M, Červeň J, Oppelt J, Peteja M, Vávra P, Zonča P, Procházka V, Brázda V, Pečinka P. Liver regeneration during the associating liver partition and portal vein ligation for staged hepatectomy procedure in Sus scrofa is positively modulated by stem cells. Oncol Lett 2018; 15:6309-6321. [PMID: 29616108 PMCID: PMC5876427 DOI: 10.3892/ol.2018.8108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 11/02/2017] [Indexed: 11/17/2022] Open
Abstract
This present study investigated the impact of the application of stem cells to liver regeneration following the first stage of associating liver partition and portal vein ligation for staged hepatectomy (ALPPS). The experiment was conducted on a pig model (n=6, 3 that did not receive application of stem cells, 3 that received application stem cells). Collected samples of liver (day 0 and 9 following surgery) were subjected to complete transcriptome sequencing. In total, 39 differentially expressed genes were found in the group without the application of the stem cells (genes of unwanted processes such as fibrosis and inflammation). In the group that did receive application of stem cells, no significantly differentially expressed genes were found, indicating a properly regenerated liver remnant. The present study therefore demonstrated, to the best of our knowledge for the first time, the positive effect of stem cells application in the liver regeneration process during ALPPS procedure in the pig model.
Collapse
Affiliation(s)
- Martin Bartas
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, 71000 Ostrava, Czech Republic
| | - Jiri Červeň
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, 71000 Ostrava, Czech Republic.,Institute of Environmental Technologies, Faculty of Science, University of Ostrava, 71000 Ostrava, Czech Republic
| | - Jan Oppelt
- Centre for Structural Biology, Central European Institute of Technology, Masaryk University, 62500 Brno, Czech Republic.,National Centre for Biomolecular Research, Centre for Structural Biology, Central European Institute of Technology, Masaryk University, 62500 Brno, 70852 Ostrava, Czech Republic
| | - Matus Peteja
- Department of Surgery, University Hospital in Ostrava, 70852 Ostrava, Czech Republic.,Department of Surgical Studies, Faculty of Medicine, University of Ostrava, 70852 Ostrava, Czech Republic
| | - Petr Vávra
- Department of Surgery, University Hospital in Ostrava, 70852 Ostrava, Czech Republic.,Department of Surgical Studies, Faculty of Medicine, University of Ostrava, 70852 Ostrava, Czech Republic
| | - Pavel Zonča
- Department of Surgery, University Hospital in Ostrava, 70852 Ostrava, Czech Republic.,Department of Surgical Studies, Faculty of Medicine, University of Ostrava, 70852 Ostrava, Czech Republic
| | - Vaclav Procházka
- Department of Radiology, University Hospital in Ostrava, 70852 Ostrava, Czech Republic
| | - Vaclav Brázda
- Institute of Biophysics, Academy of Sciences of The Czech Republic, 61265 Brno, Czech Republic
| | - Petr Pečinka
- Department of Biology and Ecology, Faculty of Science, University of Ostrava, 71000 Ostrava, Czech Republic.,Institute of Environmental Technologies, Faculty of Science, University of Ostrava, 71000 Ostrava, Czech Republic
| |
Collapse
|
65
|
Interleukin-4 and interleukin-13 increase NADPH oxidase 1-related proliferation of human colon cancer cells. Oncotarget 2018; 8:38113-38135. [PMID: 28498822 PMCID: PMC5503519 DOI: 10.18632/oncotarget.17494] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/17/2017] [Indexed: 01/01/2023] Open
Abstract
Human colon cancers express higher levels of NADPH oxidase 1 [NOX1] than adjacent normal epithelium. It has been suggested that reactive oxygen species [ROS] derived from NOX1 contribute to DNA damage and neoplastic transformation in the colon, particularly during chronic inflammatory stress. However, the mechanism(s) underlying increased NOX1 expression in malignant tumors or chronic inflammatory states involving the intestine are poorly characterized. We examined the effects of two pro-inflammatory cytokines, IL-4 and IL-13, on the regulation of NOX1. NOX1 expression was increased 4- to 5-fold in a time- and concentration-dependent manner by both cytokines in human colon cancer cell lines when a functional Type II IL-4 receptor was present. Increased NOX1 transcription following IL-4/IL-13 exposure was mediated by JAK1/STAT6 signaling, was associated with a ROS-related inhibition of protein tyrosine phosphatase activity, and was dependent upon activation and specific binding of GATA3 to the NOX1 promoter. NOX1-mediated ROS production increased cell cycle progression through S-phase leading to a significant increase in cellular proliferation. Evaluation of twenty pairs of surgically-resected colon cancers and their associated uninvolved adjacent colonic epithelium demonstrated a significant increase in the active form of NOX1, NOX1-L, in tumors compared to normal tissues, and a significant correlation between the expression levels of NOX1 and the Type II IL-4 receptor in tumor and the uninvolved colon. These studies imply that NOX1 expression, mediated by IL-4/IL-13, could contribute to an oxidant milieu capable of supporting the initiation or progression of colonic cancer, suggesting a role for NOX1 as a therapeutic target.
Collapse
|
66
|
Insights on Localized and Systemic Delivery of Redox-Based Therapeutics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2468457. [PMID: 29636836 PMCID: PMC5832094 DOI: 10.1155/2018/2468457] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022]
Abstract
Reactive oxygen and nitrogen species are indispensable in cellular physiology and signaling. Overproduction of these reactive species or failure to maintain their levels within the physiological range results in cellular redox dysfunction, often termed cellular oxidative stress. Redox dysfunction in turn is at the molecular basis of disease etiology and progression. Accordingly, antioxidant intervention to restore redox homeostasis has been pursued as a therapeutic strategy for cardiovascular disease, cancer, and neurodegenerative disorders among many others. Despite preliminary success in cellular and animal models, redox-based interventions have virtually been ineffective in clinical trials. We propose the fundamental reason for their failure is a flawed delivery approach. Namely, systemic delivery for a geographically local disease limits the effectiveness of the antioxidant. We take a critical look at the literature and evaluate successful and unsuccessful approaches to translation of redox intervention to the clinical arena, including dose, patient selection, and delivery approach. We argue that when interpreting a failed antioxidant-based clinical trial, it is crucial to take into account these variables and importantly, whether the drug had an effect on the redox status. Finally, we propose that local and targeted delivery hold promise to translate redox-based therapies from the bench to the bedside.
Collapse
|
67
|
Abstract
Tyrosine kinase substrate (Tks) adaptor proteins are considered important regulators of various physiological and/or pathological processes, particularly cell migration and invasion, and cancer progression. These proteins contain PX and SH3 domains, and act as scaffolds, bringing membrane and cellular components in close proximity in structures known as invadopodia or podosomes. Tks proteins, analogous to the related proteins p47phox, p40phox and NoxO1, also facilitate local generation of reactive oxygen species (ROS), which aid in signaling at invadopodia and/or podosomes to promote their activity. As their name suggests, Tks adaptor proteins are substrates for tyrosine kinases, especially Src. In this Cell Science at a Glance article and accompanying poster, we discuss the known structural and functional aspects of Tks adaptor proteins. As the science of Tks proteins is evolving, this article will point out where we stand and what still needs to be explored. We also underscore pathological conditions involving these proteins, providing a basis for future research to develop therapies for treatment of these diseases.
Collapse
Affiliation(s)
- Priyanka Saini
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Sara A Courtneidge
- Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
68
|
Genna A, Lapetina S, Lukic N, Twafra S, Meirson T, Sharma VP, Condeelis JS, Gil-Henn H. Pyk2 and FAK differentially regulate invadopodia formation and function in breast cancer cells. J Cell Biol 2017; 217:375-395. [PMID: 29133485 PMCID: PMC5748976 DOI: 10.1083/jcb.201702184] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 08/13/2017] [Accepted: 09/27/2017] [Indexed: 12/11/2022] Open
Abstract
The nonreceptor tyrosine kinase Pyk2 is highly expressed in invasive breast cancer, but how it potentiates tumor cell invasiveness is unclear. Genna et al. find that Pyk2 and the closely related kinase FAK modulate breast cancer cell invasiveness by distinct mechanisms and coordinate the balance between focal adhesion–mediated migration and invadopodia-dependent extracellular matrix invasion. The nonreceptor tyrosine kinase Pyk2 is highly expressed in invasive breast cancer, but the mechanism by which it potentiates tumor cell invasiveness is unclear at present. Using high-throughput protein array screening and bioinformatic analysis, we identified cortactin as a novel substrate and interactor of proline-rich tyrosine kinase 2 (Pyk2). Pyk2 colocalizes with cortactin to invadopodia of invasive breast cancer cells, where it mediates epidermal growth factor–induced cortactin tyrosine phosphorylation both directly and indirectly via Src-mediated Abl-related gene (Arg) activation, leading to actin polymerization in invadopodia, extracellular matrix degradation, and tumor cell invasion. Both Pyk2 and the closely related focal adhesion kinase (FAK) regulate tumor cell invasion, albeit via distinct mechanisms. Although Pyk2 regulates tumor cell invasion by controlling invadopodium-mediated functions, FAK controls invasiveness of tumor cells by regulating focal adhesion–mediated motility. Collectively, our findings identify Pyk2 as a unique mediator of invadopodium formation and function and also provide a novel insight into the mechanisms by which Pyk2 mediates tumor cell invasion.
Collapse
Affiliation(s)
- Alessandro Genna
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | | | - Nikola Lukic
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Shams Twafra
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Tomer Meirson
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Ved P Sharma
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY.,Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY.,Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY
| | - John S Condeelis
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY.,Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY.,Integrated Imaging Program, Albert Einstein College of Medicine, Bronx, NY
| | - Hava Gil-Henn
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
69
|
Rodic S, Vincent MD. Reactive oxygen species (ROS) are a key determinant of cancer's metabolic phenotype. Int J Cancer 2017; 142:440-448. [PMID: 28940517 DOI: 10.1002/ijc.31069] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/28/2017] [Accepted: 09/11/2017] [Indexed: 12/21/2022]
Abstract
Cancer cells exhibit a wide range of metabolic phenotypes, ranging from strict aerobic glycolysis to increased mitochondrial respiration. The cause and utility of this metabolic variation is poorly understood. Given that cancer cells experience heavy selection within their microenvironment, survival requires metabolic adaptation to both extracellular and intracellular conditions. Herein, we suggest that reactive oxygen species (ROS) are a key determinant of cancer's metabolic phenotype. Intracellular ROS levels can be modified by an assortment of critical parameters including oxygenation, glucose availability and growth factors. ROS act as integrators of environmental information as well as downstream effectors of signaling pathways. Maintaining ROS within a narrow range allows malignant cells to enhance growth and invasion while limiting their apoptotic susceptibility. Cancer cells actively modify their metabolism to optimize intracellular ROS levels and thereby improve survival. Furthermore, we highlight distinct metabolic phenotypes in response to oxidative stress and their tumorigenic drivers.
Collapse
Affiliation(s)
- Stefan Rodic
- Schulich School of Medicine and Dentistry, 1151 Richmond St, Western University, London, ON, Canada
| | - Mark David Vincent
- Schulich School of Medicine and Dentistry, 1151 Richmond St, Western University, London, ON, Canada.,Department of Medical Oncology, London Regional Cancer Program, 800 Commissioners Road East, London, ON, Canada
| |
Collapse
|
70
|
Meitzler JL, Makhlouf HR, Antony S, Wu Y, Butcher D, Jiang G, Juhasz A, Lu J, Dahan I, Jansen-Dürr P, Pircher H, Shah AM, Roy K, Doroshow JH. Decoding NADPH oxidase 4 expression in human tumors. Redox Biol 2017; 13:182-195. [PMID: 28578276 PMCID: PMC5458090 DOI: 10.1016/j.redox.2017.05.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 05/23/2017] [Indexed: 12/27/2022] Open
Abstract
NADPH oxidase 4 (NOX4) is a redox active, membrane-associated protein that contributes to genomic instability, redox signaling, and radiation sensitivity in human cancers based on its capacity to generate H2O2 constitutively. Most studies of NOX4 in malignancy have focused on the evaluation of a small number of tumor cell lines and not on human tumor specimens themselves; furthermore, these studies have often employed immunological tools that have not been well characterized. To determine the prevalence of NOX4 expression across a broad range of solid tumors, we developed a novel monoclonal antibody that recognizes a specific extracellular region of the human NOX4 protein, and that does not cross-react with any of the other six members of the NOX gene family. Evaluation of 20 sets of epithelial tumors revealed, for the first time, high levels of NOX4 expression in carcinomas of the head and neck (15/19 patients), esophagus (12/18 patients), bladder (10/19 patients), ovary (6/17 patients), and prostate (7/19 patients), as well as malignant melanoma (7/15 patients) when these tumors were compared to histologically-uninvolved specimens from the same organs. Detection of NOX4 protein upregulation by low levels of TGF-β1 demonstrated the sensitivity of this new probe; and immunofluorescence experiments found that high levels of endogenous NOX4 expression in ovarian cancer cells were only demonstrable associated with perinuclear membranes. These studies suggest that NOX4 expression is upregulated, compared to normal tissues, in a well-defined, and specific group of human carcinomas, and that its expression is localized on intracellular membranes in a fashion that could modulate oxidative DNA damage.
Collapse
Affiliation(s)
- Jennifer L Meitzler
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Hala R Makhlouf
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Smitha Antony
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yongzhong Wu
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Donna Butcher
- Pathology/Histotechnology Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, NIH, Frederick, MD 21702, USA
| | - Guojian Jiang
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Agnes Juhasz
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jiamo Lu
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Iris Dahan
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, 6020 Innsbruck, Austria
| | - Haymo Pircher
- Institute for Biomedical Aging Research and Center for Molecular Biosciences Innsbruck (CMBI), Universität Innsbruck, 6020 Innsbruck, Austria
| | - Ajay M Shah
- King's College London British Heart Foundation Centre, Cardiovascular Division, James Black Centre, London SE5 9NU, United Kingdom
| | - Krishnendu Roy
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - James H Doroshow
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA; Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
71
|
Little AC, Sulovari A, Danyal K, Heppner DE, Seward DJ, van der Vliet A. Paradoxical roles of dual oxidases in cancer biology. Free Radic Biol Med 2017; 110:117-132. [PMID: 28578013 PMCID: PMC5535817 DOI: 10.1016/j.freeradbiomed.2017.05.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 02/06/2023]
Abstract
Dysregulated oxidative metabolism is a well-recognized aspect of cancer biology, and many therapeutic strategies are based on targeting cancers by altering cellular redox pathways. The NADPH oxidases (NOXes) present an important enzymatic source of biological oxidants, and the expression and activation of several NOX isoforms are frequently dysregulated in many cancers. Cell-based studies have demonstrated a role for several NOX isozymes in controlling cell proliferation and/or cell migration, further supporting a potential contributing role for NOX in promoting cancer. While various NOX isoforms are often upregulated in cancers, paradoxical recent findings indicate that dual oxidases (DUOXes), normally prominently expressed in epithelial lineages, are frequently suppressed in epithelial-derived cancers by epigenetic mechanisms, although the functional relevance of such DUOX silencing has remained unclear. This review will briefly summarize our current understanding regarding the importance of reactive oxygen species (ROS) and NOXes in cancer biology, and focus on recent observations indicating the unique and seemingly opposing roles of DUOX enzymes in cancer biology. We will discuss current knowledge regarding the functional properties of DUOX, and recent studies highlighting mechanistic consequences of DUOX1 loss in lung cancer, and its consequences for tumor invasiveness and current anticancer therapy. Finally, we will also discuss potentially unique roles for the DUOX maturation factors. Overall, a better understanding of mechanisms that regulate DUOX and the functional consequences of DUOX silencing in cancer may offer valuable new diagnostic insights and novel therapeutic opportunities.
Collapse
Affiliation(s)
- Andrew C Little
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States
| | - Arvis Sulovari
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States; Department of Microbiology and Molecular Genetics, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - Karamatullah Danyal
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - David E Heppner
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - David J Seward
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, United States; Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405, United States.
| |
Collapse
|
72
|
Aydin E, Johansson J, Nazir FH, Hellstrand K, Martner A. Role of NOX2-Derived Reactive Oxygen Species in NK Cell-Mediated Control of Murine Melanoma Metastasis. Cancer Immunol Res 2017; 5:804-811. [PMID: 28760732 DOI: 10.1158/2326-6066.cir-16-0382] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 05/04/2017] [Accepted: 07/20/2017] [Indexed: 11/16/2022]
Abstract
The NADPH oxidase of myeloid cells, NOX2, generates reactive oxygen species (ROS) to eliminate pathogens and malignant cells. NOX2-derived ROS have also been proposed to dampen functions of natural killer (NK) cells and other antineoplastic lymphocytes in the microenvironment of established tumors. The mechanisms by which NOX2 and ROS influence the process of distant metastasis have only been partially explored. Here, we utilized genetically NOX2-deficient mice and pharmacologic inhibition of NOX2 to elucidate the role of NOX2 for the hematogenous metastasis of melanoma cells. After intravenous inoculation of B16F1 or B16F10 cells, lung metastasis formation was reduced in B6.129S6-Cybbtm1DinK (Nox2-KO) versus Nox2-sufficient wild-type (WT) mice. Systemic treatment with the NOX2-inhibitor histamine dihydrochloride (HDC) reduced melanoma metastasis and enhanced the infiltration of IFNγ-producing NK cells into lungs of WT but not of Nox2-KO mice. IFNγ-deficient B6.129S7-Ifngtm1Ts /J mice were prone to develop melanoma metastases and did not respond to in vivo treatment with HDC. We propose that NOX2-derived ROS facilitate metastasis of melanoma cells by downmodulating NK-cell function and that inhibition of NOX2 may restore IFNγ-dependent, NK cell-mediated clearance of melanoma cells. Cancer Immunol Res; 5(9); 804-11. ©2017 AACR.
Collapse
Affiliation(s)
- Ebru Aydin
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Junko Johansson
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Faisal Hayat Nazir
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Kristoffer Hellstrand
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Anna Martner
- TIMM Laboratory, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
73
|
Paterson EK, Courtneidge SA. Invadosomes are coming: new insights into function and disease relevance. FEBS J 2017; 285:8-27. [PMID: 28548369 DOI: 10.1111/febs.14123] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/09/2017] [Accepted: 05/24/2017] [Indexed: 12/21/2022]
Abstract
Invadopodia and podosomes are discrete, actin-based molecular protrusions that form in cancer cells and normal cells, respectively, in response to diverse signaling pathways and extracellular matrix cues. Although they participate in a host of different cellular processes, they share a common functional theme of controlling pericellular proteolytic activity, which sets them apart from other structures that function in migration and adhesion, including focal adhesions, lamellipodia, and filopodia. In this review, we highlight research that explores the function of these complex structures, including roles for podosomes in embryonic and postnatal development, in angiogenesis and remodeling of the vasculature, in maturation of the postsynaptic membrane, in antigen sampling and recognition, and in cell-cell fusion mechanisms, as well as the involvement of invadopodia at multiple steps of the metastatic cascade, and how all of this may apply in the treatment of human disease states. Finally, we explore recent research that implicates a novel role for exosomes and microvesicles in invadopodia-dependent and invadopodia-independent mechanisms of invasion, respectively.
Collapse
Affiliation(s)
- Elyse K Paterson
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Sara A Courtneidge
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
74
|
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: 98] [Impact Index Per Article: 12.3] [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
|
75
|
Abstract
Abstract
RHAMM is hyaluronan- receptor with multiple functions in the cell, RHAMM is involved in proliferation, motility, migration, invasion, mitotic spindle formation in tumour cells. Therefore, RHAMM could be a relevant target for molecular targeted therapies against tumors.The role of RHAMM-target peptides in inhibition invasion for preventing breast cancer has not yet been investigated. Base on this, we analyzed the RHAMM-target peptides for their therapeutic activity against breast cancer cells. In the present study, we examined the effect of RHAMM-target peptides on the invasion of breast cancer cells (MDAMB- 231), using confocal microscopy. We shown that RHAMM-target peptides decreased formation of invadopodia of breast cancer cells. The treatment of breast cancer cells by RHAMM -target peptides inhibited the invasion up to 99 %. Additionally, RHAMM-target peptides induced the morphological changes of of breast cancer cells. Therefore, based on these results, we can conclude that RHAMM-target peptides may be potential anti-cancer agents.
Collapse
Affiliation(s)
- Natalia Akentieva
- Kinetics Chemical and Biological Processes; Institute problems of Chemical Physics RAS, pr.acad. Semenova, 1 Chernogolovka Moscow , Russian Federation
| |
Collapse
|
76
|
Chio IIC, Tuveson DA. ROS in Cancer: The Burning Question. Trends Mol Med 2017; 23:411-429. [PMID: 28427863 PMCID: PMC5462452 DOI: 10.1016/j.molmed.2017.03.004] [Citation(s) in RCA: 370] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 02/07/2023]
Abstract
An unanswered question in human health is whether antioxidation prevents or promotes cancer. Antioxidation has historically been viewed as chemopreventive, but emerging evidence suggests that antioxidants may be supportive of neoplasia. We posit this contention to be rooted in the fact that ROS do not operate as one single biochemical entity, but as diverse secondary messengers in cancer cells. This cautions against therapeutic strategies to increase ROS at a global level. To leverage redox alterations towards the development of effective therapies necessitates the application of biophysical and biochemical approaches to define redox dynamics and to functionally elucidate specific oxidative modifications in cancer versus normal cells. An improved understanding of the sophisticated workings of redox biology is imperative to defeating cancer.
Collapse
Affiliation(s)
- Iok In Christine Chio
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA.
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
77
|
Morry J, Ngamcherdtrakul W, Yantasee W. Oxidative stress in cancer and fibrosis: Opportunity for therapeutic intervention with antioxidant compounds, enzymes, and nanoparticles. Redox Biol 2017; 11:240-253. [PMID: 28012439 PMCID: PMC5198743 DOI: 10.1016/j.redox.2016.12.011] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 12/05/2016] [Accepted: 12/06/2016] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress, mainly contributed by reactive oxygen species (ROS), has been implicated in pathogenesis of several diseases. We review two primary examples; fibrosis and cancer. In fibrosis, ROS promote activation and proliferation of fibroblasts and myofibroblasts, activating TGF-β pathway in an autocrine manner. In cancer, ROS account for its genomic instability, resistance to apoptosis, proliferation, and angiogenesis. Importantly, ROS trigger cancer cell invasion through invadopodia formation as well as extravasation into a distant metastasis site. Use of antioxidant supplements, enzymes, and inhibitors for ROS-generating NADPH oxidases (NOX) is a logical therapeutic intervention for fibrosis and cancer. We review such attempts, progress, and challenges. Lastly, we review how nanoparticles with inherent antioxidant activity can also be a promising therapeutic option, considering their additional feature as a delivery platform for drugs, genes, and imaging agents.
Collapse
Affiliation(s)
- Jingga Morry
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Worapol Ngamcherdtrakul
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA; PDX Pharmaceuticals, LLC, 3303 SW Bond Ave, Portland, OR 97239, USA
| | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Ave, Portland, OR 97239, USA; PDX Pharmaceuticals, LLC, 3303 SW Bond Ave, Portland, OR 97239, USA.
| |
Collapse
|
78
|
Jones RM, Neish AS. Redox signaling mediated by the gut microbiota. Free Radic Biol Med 2017; 105:41-47. [PMID: 27989756 DOI: 10.1016/j.freeradbiomed.2016.10.495] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 12/27/2022]
Abstract
The microbiota that inhabits the mammalian intestine can influence a range of physiological functions, including the modulation of immune responses, enhancement epithelial barrier function, and the stimulation of cell proliferation. While the mechanisms by which commensal prokaryotes stimulate immune signaling networks are well-characterized, less is known about the mechanistic control over homeostatic pathways within tissues. Recent reports by our research group have demonstrated that contact between the gut epithelia and some groups of enteric commensal bacteria prompts the rapid generation of reactive oxygen species (ROS) within host cells. Whereas the bacterial-induced production of ROS in phagocytes in response to ligand binding to Formyl Peptide Receptors (FPRs) and ensuing activation of NADPH oxidase 2 (Nox2) is a well-defined mechanism, ROS generated by other cell types such as intestinal epithelia in response to microbial signals via FPRs and the NADPH oxidase 1 (Nox1) is less appreciated. Importantly, enzymatically generated ROS have been shown to function as second messengers in many signal transduction pathways via the transient oxidative activity on sensor proteins bearing oxidant-sensitive thiol groups. Examples of redox sensitive proteins include tyrosine phosphatases that serve as regulators of MAPK pathways, focal adhesion kinase, as well as components involved NF-kB activation. Here, we review the leading edge discoveries gleaned from investigations that focus on microbial-induced generation of ROS and their functional effects on host physiology. These studies identify the functional molecular elements and mechanistic events that mediate the established effects of the normal microbiota on intestinal physiology.
Collapse
Affiliation(s)
- Rheinallt M Jones
- Department of Pediatrics, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michaels St, Room 105-L, Atlanta, GA 30322, United States
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michaels St, Room 105-L, Atlanta, GA 30322, United States.
| |
Collapse
|
79
|
Morry J, Ngamcherdtrakul W, Gu S, Reda M, Castro DJ, Sangvanich T, Gray JW, Yantasee W. Targeted Treatment of Metastatic Breast Cancer by PLK1 siRNA Delivered by an Antioxidant Nanoparticle Platform. Mol Cancer Ther 2017; 16:763-772. [PMID: 28138033 PMCID: PMC5445934 DOI: 10.1158/1535-7163.mct-16-0644] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/13/2016] [Accepted: 12/18/2016] [Indexed: 11/16/2022]
Abstract
Metastatic breast cancer is developed in about 20% to 30% of newly diagnosed patients with early-stage breast cancer despite treatments. Herein, we report a novel nanoparticle platform with intrinsic antimetastatic properties for the targeted delivery of Polo-like kinase 1 siRNA (siPLK1). We first evaluated it in a triple-negative breast cancer (TNBC) model, which shows high metastatic potential. PLK1 was identified as the top therapeutic target for TNBC cells and tumor-initiating cells in a kinome-wide screen. The platform consists of a 50-nm mesoporous silica nanoparticle (MSNP) core coated layer-by-layer with bioreducible cross-linked PEI and PEG polymers, conjugated with an antibody for selective uptake into cancer cells. siRNA is loaded last and fully protected under the PEG layer from blood enzymatic degradation. The material has net neutral charge and low nonspecific cytotoxicity. We have also shown for the first time that the MSNP itself inhibited cancer migration and invasion in TNBC cells owing to its ROS- and NOX4-modulating properties. In vivo, siPLK1 nanoconstructs (six doses of 0.5 mg/kg) knocked down about 80% of human PLK1 mRNA expression in metastatic breast cancer cells residing in mouse lungs and reduced tumor incidence and burden in lungs and other organs of an experimental metastasis mouse model. Long-term treatment significantly delayed the onset of death in mice and improved the overall survival. The platform capable of simultaneously inhibiting the proliferative and metastatic hallmarks of cancer progression is unique and has great therapeutic potential to also target other metastatic cancers beyond TNBC. Mol Cancer Ther; 16(4); 763-72. ©2017 AACR.
Collapse
Affiliation(s)
- Jingga Morry
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Worapol Ngamcherdtrakul
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
- PDX Pharmaceuticals, LLC, Portland, Oregon
| | - Shenda Gu
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Moataz Reda
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - David J Castro
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
- PDX Pharmaceuticals, LLC, Portland, Oregon
| | - Thanapon Sangvanich
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon
| | - Joe W Gray
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon.
| | - Wassana Yantasee
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon.
- PDX Pharmaceuticals, LLC, Portland, Oregon
| |
Collapse
|
80
|
Abstract
Reactive oxygen species (ROS), now appreciated for their cellular signaling capabilities, have a dual role in cancer. On the one hand, ROS can promote protumorigenic signaling, facilitating cancer cell proliferation, survival, and adaptation to hypoxia. On the other hand, ROS can promote antitumorigenic signaling and trigger oxidative stress-induced cancer cell death. To hyperactivate the cell signaling pathways necessary for cellular transformation and tumorigenesis, cancer cells increase their rate of ROS production compared with normal cells. Concomitantly, in order to maintain ROS homeostasis and evade cell death, cancer cells increase their antioxidant capacity. Compared with normal cells, this altered redox environment of cancer cells may increase their susceptibility to ROS-manipulation therapies. In this review, we discuss the two faces of ROS in cancer, the potential mechanisms underlying ROS signaling, and the opposing cancer therapeutic approaches to targeting ROS.
Collapse
Affiliation(s)
- Colleen R Reczek
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Navdeep S Chandel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| |
Collapse
|
81
|
Hempel N, Trebak M. Crosstalk between calcium and reactive oxygen species signaling in cancer. Cell Calcium 2017; 63:70-96. [PMID: 28143649 DOI: 10.1016/j.ceca.2017.01.007] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/13/2017] [Accepted: 01/14/2017] [Indexed: 02/07/2023]
Abstract
The interplay between Ca2+ and reactive oxygen species (ROS) signaling pathways is well established, with reciprocal regulation occurring at a number of subcellular locations. Many Ca2+ channels at the cell surface and intracellular organelles, including the endoplasmic reticulum and mitochondria are regulated by redox modifications. In turn, Ca2+ signaling can influence the cellular generation of ROS, from sources such as NADPH oxidases and mitochondria. This relationship has been explored in great depth during the process of apoptosis, where surges of Ca2+ and ROS are important mediators of cell death. More recently, coordinated and localized Ca2+ and ROS transients appear to play a major role in a vast variety of pro-survival signaling pathways that may be crucial for both physiological and pathophysiological functions. While much work is required to firmly establish this Ca2+-ROS relationship in cancer, existing evidence from other disease models suggests this crosstalk is likely of significant importance in tumorigenesis. In this review, we describe the regulation of Ca2+ channels and transporters by oxidants and discuss the potential consequences of the ROS-Ca2+ interplay in tumor cells.
Collapse
Affiliation(s)
- Nadine Hempel
- Department of Pharmacology, Penn State College of Medicine, Hershey PA 17033, United States; Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey PA 17033, United States.
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey PA 17033, United States; Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey PA 17033, United States.
| |
Collapse
|
82
|
Ma MW, Wang J, Zhang Q, Wang R, Dhandapani KM, Vadlamudi RK, Brann DW. NADPH oxidase in brain injury and neurodegenerative disorders. Mol Neurodegener 2017; 12:7. [PMID: 28095923 PMCID: PMC5240251 DOI: 10.1186/s13024-017-0150-7] [Citation(s) in RCA: 309] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress is a common denominator in the pathology of neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and multiple sclerosis, as well as in ischemic and traumatic brain injury. The brain is highly vulnerable to oxidative damage due to its high metabolic demand. However, therapies attempting to scavenge free radicals have shown little success. By shifting the focus to inhibit the generation of damaging free radicals, recent studies have identified NADPH oxidase as a major contributor to disease pathology. NADPH oxidase has the primary function to generate free radicals. In particular, there is growing evidence that the isoforms NOX1, NOX2, and NOX4 can be upregulated by a variety of neurodegenerative factors. The majority of recent studies have shown that genetic and pharmacological inhibition of NADPH oxidase enzymes are neuroprotective and able to reduce detrimental aspects of pathology following ischemic and traumatic brain injury, as well as in chronic neurodegenerative disorders. This review aims to summarize evidence supporting the role of NADPH oxidase in the pathology of these neurological disorders, explores pharmacological strategies of targeting this major oxidative stress pathway, and outlines obstacles that need to be overcome for successful translation of these therapies to the clinic.
Collapse
Affiliation(s)
- Merry W Ma
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Jing Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ruimin Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Krishnan M Dhandapani
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, 7703 Medical Drive, San Antonio, TX, 78229, USA
| | - Darrell W Brann
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA. .,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA.
| |
Collapse
|
83
|
Peuchant E, Bats ML, Moranvillier I, Lepoivre M, Guitton J, Wendum D, Lacombe ML, Moreau-Gaudry F, Boissan M, Dabernat S. Metastasis suppressor NM23 limits oxidative stress in mammals by preventing activation of stress-activated protein kinases/JNKs through its nucleoside diphosphate kinase activity. FASEB J 2017; 31:1531-1546. [PMID: 28077425 DOI: 10.1096/fj.201600705r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 12/19/2016] [Indexed: 11/11/2022]
Abstract
NME1 (nonmetastatic expressed 1) gene, which encodes nucleoside diphosphate kinase (NDPK) A [also known as nonmetastatic clone 23 (NM23)-H1 in humans and NM23-M1 in mice], is a suppressor of metastasis, but several lines of evidence-mostly from plants-also implicate it in the regulation of the oxidative stress response. Here, our aim was to investigate the physiologic relevance of NDPK A with respect to the oxidative stress response in mammals and to study its molecular basis. NME1-knockout mice died sooner, suffered greater hepatocyte injury, and had lower superoxide dismutase activity than did wild-type (WT) mice in response to paraquat-induced acute oxidative stress. Deletion of NME1 reduced total NDPK activity and exacerbated activation of the stress-related MAPK, JNK, in the liver in response to paraquat. In a mouse transformed hepatocyte cell line and in primary cultures of normal human keratinocytes, MAPK activation in response to H2O2 and UVB, respectively, was dampened by expression of NM23-M1/NM23-H1, dependent on its NDPK catalytic activity. Furthermore, excess or depletion of NM23-M1/NM23-H1 NDPK activity did not affect the intracellular bulk concentration of nucleoside di- and triphosphates. NME1-deficient mouse embryo fibroblasts grew poorly in culture, were more sensitive to stress than WT fibroblasts, and did not immortalize, which suggested that they senesce earlier than do WT fibroblasts. Collectively, these results indicate that the NDPK activity of NM23-M1/NM23-H1 protects cells from acute oxidative stress by inhibiting activation of JNK in mammal models.-Peuchant, E., Bats, M.-L., Moranvillier, I., Lepoivre, M., Guitton, J., Wendum, D., Lacombe, M.-L., Moreau-Gaudry, F., Boissan, M., Dabernat, S. Metastasis suppressor NM23 limits oxidative stress in mammals by preventing activation of stress-activated protein kinases/JNKs through its nucleoside diphosphate kinase activity.
Collapse
Affiliation(s)
- Evelyne Peuchant
- Collège Santé Université de Bordeaux, Bordeaux, France.,INSERM 1035, Bordeaux, France.,Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Marie-Lise Bats
- Collège Santé Université de Bordeaux, Bordeaux, France.,Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Isabelle Moranvillier
- Collège Santé Université de Bordeaux, Bordeaux, France.,INSERM 1035, Bordeaux, France
| | - Michel Lepoivre
- Université Paris Sud, Commissariat à l'Énergie Atomique et aux Énergies, Unité Mixte de Recherche, Centre National de la Recherche Scientifique 9198, Orsay, France
| | - Jérôme Guitton
- Hospices Civils de Lyon, Pierre Bénite, France.,Université de Lyon, Lyon, France
| | - Dominique Wendum
- Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, INSERM, Unité Mixte de Recherche S938, Saint-Antoine Research Center, Paris, France.,Laboratoire d'Anatomie Pathologique, Hôpital Saint-Antoine, Paris, France
| | - Marie-Lise Lacombe
- Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, INSERM, Unité Mixte de Recherche S938, Saint-Antoine Research Center, Paris, France
| | - François Moreau-Gaudry
- Collège Santé Université de Bordeaux, Bordeaux, France.,INSERM 1035, Bordeaux, France.,Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Mathieu Boissan
- Sorbonne Universités, Université Pierre et Marie Curie, Université Paris 06, INSERM, Unité Mixte de Recherche S938, Saint-Antoine Research Center, Paris, France; .,Service de Biochimie et Hormonologie, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Sandrine Dabernat
- Collège Santé Université de Bordeaux, Bordeaux, France; .,INSERM 1035, Bordeaux, France.,Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| |
Collapse
|
84
|
Ferreira LF, Laitano O. Regulation of NADPH oxidases in skeletal muscle. Free Radic Biol Med 2016; 98:18-28. [PMID: 27184955 PMCID: PMC4975970 DOI: 10.1016/j.freeradbiomed.2016.05.011] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/31/2016] [Accepted: 05/12/2016] [Indexed: 12/20/2022]
Abstract
The only known function of NAD(P)H oxidases is to produce reactive oxygen species (ROS). Skeletal muscles express three isoforms of NAD(P)H oxidases (Nox1, Nox2, and Nox4) that have been identified as critical modulators of redox homeostasis. Nox2 acts as the main source of skeletal muscle ROS during contractions, participates in insulin signaling and glucose transport, and mediates the myocyte response to osmotic stress. Nox2 and Nox4 contribute to skeletal muscle abnormalities elicited by angiotensin II, muscular dystrophy, heart failure, and high fat diet. Our review addresses the expression and regulation of NAD(P)H oxidases with emphasis on aspects that are relevant to skeletal muscle. We also summarize: i) the most widely used NAD(P)H oxidases activity assays and inhibitors, and ii) studies that have defined Nox enzymes as protagonists of skeletal muscle redox homeostasis in a variety of health and disease conditions.
Collapse
Affiliation(s)
- Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA.
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Universidade Federal do Vale do São Francisco, Petrolina, PE, Brazil
| |
Collapse
|
85
|
Logun MT, Bisel NS, Tanasse EA, Zhao W, Gunasekera B, Mao L, Karumbaiah L. Glioma Cell Invasion is Significantly Enhanced in Composite Hydrogel Matrices Composed of Chondroitin 4- and 4,6-Sulfated Glycosaminoglycans. J Mater Chem B 2016; 4:6052-6064. [PMID: 28217304 DOI: 10.1039/c6tb01083k] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive form of astrocytoma accounting for a majority of primary malignant brain tumors in the United States. Chondroitin sulfate proteoglycans (CSPGs) and their glycosaminoglycan (GAG) side chains are key constituents of the brain extracellular matrix (ECM) implicated in promoting tumor invasion. However, the mechanisms by which sulfated CS-GAGs promote brain tumor invasion are currently unknown. We hypothesize that glioma cell invasion is triggered by the altered sulfation of CS-GAGs in the tumor extracellular environment, and that this is potentially mediated by independent mechanisms involving CXCL12/CXCR4 and LAR signaling respectively. This was tested in vitro by encapsulating the human glioma cell line U87MG-EGFP into monosulfated (4-sulfated; CS-A), composite (4 and 4,6-sulfated; CS-A/E), unsulfated hyaluronic acid (HA), and unsulfated agarose (AG; polysaccharide) hydrogels within microfluidics-based choice assays. Our results demonstrated the enhanced preferential cell invasion into composite hydrogels, when compared to other hydrogel matrices (p<0.05). Haptotaxis assays demonstrated the significantly (p<0.05) faster migration of U87MG-EGFP cells in CXCL12 containing CS-GAG hydrogels when compared to other hydrogel matrices containing the same chemokine concentration. This is likely due to the significantly (p<0.05) greater affinity of composite CS-GAGs to CXCL12 over other hydrogel matrices. Results from qRT-PCR assays further demonstrated the significant (p<0.05) upregulation of the chemokine receptor CXCR4, and the CSPG receptor LAR in glioma cells within CS-GAG hydrogels compared to control hydrogels. Western blot analysis of cell lysates derived from glioma cells encapsulated in different hydrogel matrices further corroborate qRT-PCR results, and indicate the presence of a potential variant of LAR that is selectively expressed only in glioma cells encapsulated in CS-GAG hydrogels. These results suggest that sulfated CS-GAGs may directly induce enhanced invasion and haptotaxis of glioma cells associated with aggressive brain tumors via distinct mechanisms.
Collapse
Affiliation(s)
- Meghan T Logun
- Regenerative Bioscience Center, ADS Complex, University of Georgia, Athens, Georgia
| | - Nicole S Bisel
- Regenerative Bioscience Center, ADS Complex, University of Georgia, Athens, Georgia
| | - Emily A Tanasse
- College of Engineering, Boise State University, Boise, Idaho
| | - Wujun Zhao
- Department of Chemistry, University of Georgia, Athens, Georgia
| | - Bhagya Gunasekera
- Regenerative Bioscience Center, ADS Complex, University of Georgia, Athens, Georgia
| | - Leidong Mao
- College of Engineering, University of Georgia, Athens, Georgia
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, ADS Complex, University of Georgia, Athens, Georgia
| |
Collapse
|
86
|
Castro-Castro A, Marchesin V, Monteiro P, Lodillinsky C, Rossé C, Chavrier P. Cellular and Molecular Mechanisms of MT1-MMP-Dependent Cancer Cell Invasion. Annu Rev Cell Dev Biol 2016; 32:555-576. [PMID: 27501444 DOI: 10.1146/annurev-cellbio-111315-125227] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Metastasis is responsible for most cancer-associated deaths. Accumulating evidence based on 3D migration models has revealed a diversity of invasive migratory schemes reflecting the plasticity of tumor cells to switch between proteolytic and nonproteolytic modes of invasion. Yet, initial stages of localized regional tumor dissemination require proteolytic remodeling of the extracellular matrix to overcome tissue barriers. Recent data indicate that surface-exposed membrane type 1-matrix metalloproteinase (MT1-MMP), belonging to a group of membrane-anchored MMPs, plays a central role in pericellular matrix degradation during basement membrane and interstitial tissue transmigration programs. In addition, a large body of work indicates that MT1-MMP is targeted to specialized actin-rich cell protrusions termed invadopodia, which are responsible for matrix degradation. This review describes the multistep assembly of actin-based invadopodia in molecular details. Mechanisms underlying MT1-MMP traffic to invadopodia through endocytosis/recycling cycles, which are key to the invasive program of carcinoma cells, are discussed.
Collapse
Affiliation(s)
| | | | - Pedro Monteiro
- Barts Cancer Institute, University of London John Vane Science Centre, London EC1M 6BQ, United Kingdom
| | - Catalina Lodillinsky
- Instituto de Oncologia Ángel H. Roffo, Research Area, Buenos Aires, C1417DTB, Argentina
| | - Carine Rossé
- Institut Curie, Paris, F-75248 France; .,PSL Research University, Paris, F-75005 France.,CNRS, UMR 144, Paris, F-75248 France
| | - Philippe Chavrier
- Institut Curie, Paris, F-75248 France; .,PSL Research University, Paris, F-75005 France.,CNRS, UMR 144, Paris, F-75248 France
| |
Collapse
|
87
|
Tsai WC, Chen CL, Chen HC. Protein tyrosine phosphatase SHP2 promotes invadopodia formation through suppression of Rho signaling. Oncotarget 2016. [PMID: 26204488 PMCID: PMC4695156 DOI: 10.18632/oncotarget.4313] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Invadopodia are actin-enriched membrane protrusions that are important for extracellular matrix degradation and invasive cell motility. Src homolog domain-containing phosphatase 2 (SHP2), a non-receptor protein tyrosine phosphatase, has been shown to play an important role in promoting cancer metastasis, but the underlying mechanism is unclear. In this study, we found that depletion of SHP2 by short-hairpin RNA suppressed invadopodia formation in several cancer cell lines, particularly in the SAS head and neck squamous cell line. In contrast, overexpression of SHP2 promoted invadopodia formation in the CAL27 head and neck squamous cell line, which expresses low levels of endogenous SHP2. The depletion of SHP2 in SAS cells significantly decreased their invasive motility. The suppression of invadopodia formation by SHP2 depletion was restored by the Clostridium botulinum C3 exoenzyme (a Rho GTPase inhibitor) or Y27632 (a specific inhibitor for Rho-associated kinase). Together, our results suggest that SHP2 may promote invadopodia formation through inhibition of Rho signaling in cancer cells.
Collapse
Affiliation(s)
- Wan-Chen Tsai
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chien-Lin Chen
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.,Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.,Rong-Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Hong-Chen Chen
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan.,Institutue of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.,Rong-Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
88
|
Significance of kinase activity in the dynamic invadosome. Eur J Cell Biol 2016; 95:483-492. [PMID: 27465307 DOI: 10.1016/j.ejcb.2016.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/08/2016] [Accepted: 07/13/2016] [Indexed: 12/19/2022] Open
Abstract
Invadosomes are actin rich protrusive structures that facilitate invasive migration in multiple cell types. Comprised of invadopodia and podosomes, these highly dynamic structures adhere to and degrade the extracellular matrix, and are also thought to play a role in mechanosensing. Many extracellular signals have been implicated in invadosome stimulation, activating complex signalling cascades to drive the formation, activity and turnover of invadosomes. While the structural components of invadosomes have been well studied, the regulation of invadosome dynamics is still poorly understood. Protein kinases are essential to this regulation, affecting all stages of invadosome dynamics and allowing tight spatiotemporal control of their activity. Invadosome organisation and function have been linked to pathophysiological states such as cancer invasion and metastasis; therapeutic targeting of invadosome regulatory components is thus warranted. In this review, we discuss the involvement of kinase signalling in every stage of the invadosome life cycle and evaluate its significance.
Collapse
|
89
|
Charbonneau M, Lavoie RR, Lauzier A, Harper K, McDonald PP, Dubois CM. Platelet-Derived Growth Factor Receptor Activation Promotes the Prodestructive Invadosome-Forming Phenotype of Synoviocytes from Patients with Rheumatoid Arthritis. THE JOURNAL OF IMMUNOLOGY 2016; 196:3264-75. [DOI: 10.4049/jimmunol.1500502] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 02/15/2016] [Indexed: 11/19/2022]
|
90
|
Shneyer BI, Ušaj M, Henn A. Myo19 is an outer mitochondrial membrane motor and effector of starvation-induced filopodia. J Cell Sci 2015; 129:543-56. [PMID: 26659663 DOI: 10.1242/jcs.175349] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 12/05/2015] [Indexed: 12/13/2022] Open
Abstract
Mitochondria respond to environmental cues and stress conditions. Additionally, the disruption of the mitochondrial network dynamics and its distribution is implicated in a variety of neurodegenerative diseases. Here, we reveal a new function for Myo19 in mitochondrial dynamics and localization during the cellular response to glucose starvation. Ectopically expressed Myo19 localized with mitochondria to the tips of starvation-induced filopodia. Corollary to this, RNA interference (RNAi)-mediated knockdown of Myo19 diminished filopodia formation without evident effects on the mitochondrial network. We analyzed the Myo19-mitochondria interaction, and demonstrated that Myo19 is uniquely anchored to the outer mitochondrial membrane (OMM) through a 30-45-residue motif, indicating that Myo19 is a stably attached OMM molecular motor. Our work reveals a new function for Myo19 in mitochondrial positioning under stress.
Collapse
Affiliation(s)
- Boris I Shneyer
- Department of Biology, Technion Israel Institute of Technology, Haifa 3200003, Israel
| | - Marko Ušaj
- Department of Biology, Technion Israel Institute of Technology, Haifa 3200003, Israel
| | - Arnon Henn
- Department of Biology, Technion Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
91
|
Moodley S, Hui Bai X, Kapus A, Yang B, Liu M. XB130/Tks5 scaffold protein interaction regulates Src-mediated cell proliferation and survival. Mol Biol Cell 2015; 26:4492-502. [PMID: 26446840 PMCID: PMC4666142 DOI: 10.1091/mbc.e15-07-0483] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/29/2015] [Indexed: 12/20/2022] Open
Abstract
XB130 and Tks5 interact endogenously and form a complex with Src tyrosine kinase. Tks5, like XB130, plays a role in cell proliferation and cell survival, and the interaction between XB130 and Tks5 is critical for regulation of Src-mediated cell proliferation and survival. The scaffold protein XB130 regulates cell growth, survival, and migration. Yeast two-hybrid screening suggests that XB130 interacts with another scaffold protein, Tks5. We hypothesized that XB130 and Tks5 form a macromolecular complex to mediate signal transduction cascades for the regulation of cell growth and survival. Coimmunoprecipitation demonstrated that XB130 and Tks5 interact endogenously and form a complex with Src tyrosine kinase. Structure–function studies showed that the fifth SH3 domain of Tks5 binds to the N-terminus of XB130, which contains polyproline-rich motifs. Cell growth and survival studies revealed that down-regulation of XB130 and/or Tks5 reduced cell proliferation, resulting in cell cycle inhibition at the G1 phase and increased caspase 3 activity and apoptosis. Moreover, cell proliferation and survival were increased by overexpression of XB130 or Tks5 but decreased when XB130/Tks5 binding was disrupted by overexpression of XB130 N-terminal deleted mutant and/or Tks5 fifth SH3 domain W1108A mutant. Furthermore, down-regulation of XB130 and/or Tks5 inhibited serum- and growth factor–induced Src activation and downstream phosphorylation of PI3K and Akt. Our results suggest that Tks5, similar to XB130, plays a role in cell proliferation and cell survival and that the interaction between XB130 and Tks5 appears to be critical for regulation of Src-mediated cellular homeostasis.
Collapse
Affiliation(s)
- Serisha Moodley
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Xiao Hui Bai
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Andras Kapus
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada Department of Surgery, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Burton Yang
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Mingyao Liu
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada Department of Surgery, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
92
|
Altenhöfer S, Radermacher KA, Kleikers PWM, Wingler K, Schmidt HHHW. Evolution of NADPH Oxidase Inhibitors: Selectivity and Mechanisms for Target Engagement. Antioxid Redox Signal 2015; 23:406-27. [PMID: 24383718 PMCID: PMC4543484 DOI: 10.1089/ars.2013.5814] [Citation(s) in RCA: 408] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
SIGNIFICANCE Oxidative stress, an excess of reactive oxygen species (ROS) production versus consumption, may be involved in the pathogenesis of different diseases. The only known enzymes solely dedicated to ROS generation are nicotinamide adenine dinucleotide phosphate (NADPH) oxidases with their catalytic subunits (NOX). After the clinical failure of most antioxidant trials, NOX inhibitors are the most promising therapeutic option for diseases associated with oxidative stress. RECENT ADVANCES Historical NADPH oxidase inhibitors, apocynin and diphenylene iodonium, are un-specific and not isoform selective. Novel NOX inhibitors stemming from rational drug discovery approaches, for example, GKT137831, ML171, and VAS2870, show improved specificity for NADPH oxidases and moderate NOX isoform selectivity. Along with NOX2 docking sequence (NOX2ds)-tat, a peptide-based inhibitor, the use of these novel small molecules in animal models has provided preliminary in vivo evidence for a pathophysiological role of specific NOX isoforms. CRITICAL ISSUES Here, we discuss whether novel NOX inhibitors enable reliable validation of NOX isoforms' pathological roles and whether this knowledge supports translation into pharmacological applications. Modern NOX inhibitors have increased the evidence for pathophysiological roles of NADPH oxidases. However, in comparison to knockout mouse models, NOX inhibitors have limited isoform selectivity. Thus, their use does not enable clear statements on the involvement of individual NOX isoforms in a given disease. FUTURE DIRECTIONS The development of isoform-selective NOX inhibitors and biologicals will enable reliable validation of specific NOX isoforms in disease models other than the mouse. Finally, GKT137831, the first NOX inhibitor in clinical development, is poised to provide proof of principle for the clinical potential of NOX inhibition.
Collapse
Affiliation(s)
- Sebastian Altenhöfer
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht, the Netherlands
| | - Kim A Radermacher
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht, the Netherlands
| | - Pamela W M Kleikers
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht, the Netherlands
| | - Kirstin Wingler
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht, the Netherlands
| | - Harald H H W Schmidt
- Department of Pharmacology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht, the Netherlands
| |
Collapse
|
93
|
Veillat V, Spuul P, Daubon T, Egaña I, Kramer IJ, Génot E. Podosomes: Multipurpose organelles? Int J Biochem Cell Biol 2015; 65:52-60. [DOI: 10.1016/j.biocel.2015.05.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/20/2015] [Indexed: 01/11/2023]
|
94
|
Gould CM, Courtneidge SA. Regulation of invadopodia by the tumor microenvironment. Cell Adh Migr 2015; 8:226-35. [PMID: 24714597 DOI: 10.4161/cam.28346] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The tumor microenvironment consists of stromal cells, extracellular matrix (ECM), and signaling molecules that communicate with cancer cells. As tumors grow and develop, the tumor microenvironment changes. In addition, the tumor microenvironment is not only influenced by signals from tumor cells, but also stromal components contribute to tumor progression and metastasis by affecting cancer cell function. One of the mechanisms that cancer cells use to invade and metastasize is mediated by actin-rich, proteolytic structures called invadopodia. Here, we discuss how signals from the tumor environment, including growth factors, hypoxia, pH, metabolism, and stromal cell interactions, affect the formation and function of invadopodia to regulate cancer cell invasion and metastasis. Understanding how the tumor microenvironment affects invadopodia biology could aid in the development of effective therapeutics to target cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Christine M Gould
- Tumor Microenvironment and Metastasis Program; Cancer Center; Sanford-Burnham Medical Research Institute; La Jolla, CA USA
| | - Sara A Courtneidge
- Tumor Microenvironment and Metastasis Program; Cancer Center; Sanford-Burnham Medical Research Institute; La Jolla, CA USA
| |
Collapse
|
95
|
Ho FC, Zhang W, Li YY, Chan BP. Mechanoresponsive, omni-directional and local matrix-degrading actin protrusions in human mesenchymal stem cells microencapsulated in a 3D collagen matrix. Biomaterials 2015; 53:392-405. [DOI: 10.1016/j.biomaterials.2015.02.102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/22/2015] [Accepted: 02/24/2015] [Indexed: 10/25/2022]
|
96
|
Guo S, Chen X. The human Nox4: gene, structure, physiological function and pathological significance. J Drug Target 2015; 23:888-96. [PMID: 25950600 DOI: 10.3109/1061186x.2015.1036276] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Increased generation of reactive oxygen species (ROS) has been implicated in the pathogenesis of a variety of diseases such as cardiovascular diseases and cancer. NADPH oxidase (Nox), a multicomponent enzyme, has been identified as one of the key sources of ROS. Nox4, one of the seven members of Nox family (Nox1, Nox2, Nox3, Nox4, Nox5, Duox1 and Duox2), has been extensively investigated in recent years. Its unique structures result in the constitutive generation of hydrogen peroxide (H2O2) as the main product. As a key oxygen sensor, Nox4-derived H2O2 plays diverse roles in cell proliferation, migration and death. Increased expression of Nox4 in cancer has been observed, which participates in metastasis, angiogenesis and apoptosis. Expression of Nox4 in endothelial cells actively mediated endothelial activation, dysfunction and injury, which contributes to the development of atherosclerosis, hypertension, cardiac hypertrophy and among others. This article explores the experimental studies related to the gene, structure, physiological function and pathological significance of Nox4. As Nox4 might serve as a potential target for the therapy of cardiovascular diseases and cancer, the Nox4 inhibitor is also discussed in this article.
Collapse
Affiliation(s)
- Shuhui Guo
- a State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao , China
| | - Xiuping Chen
- a State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau , Macao , China
| |
Collapse
|
97
|
Gorin Y, Wauquier F. Upstream regulators and downstream effectors of NADPH oxidases as novel therapeutic targets for diabetic kidney disease. Mol Cells 2015; 38:285-96. [PMID: 25824546 PMCID: PMC4400302 DOI: 10.14348/molcells.2015.0010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 01/12/2015] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress has been linked to the pathogenesis of diabetic nephropathy, the complication of diabetes in the kidney. NADPH oxidases of the Nox family, and in particular the homologue Nox4, are a major source of reactive oxygen species in the diabetic kidney and are critical mediators of redox signaling in glomerular and tubulointerstitial cells exposed to the diabetic milieu. Here, we present an overview of the current knowledge related to the understanding of the role of Nox enzymes in the processes that control mesangial cell, podocyte and tubulointerstitial cell injury induced by hyperglycemia and other predominant factors enhanced in the diabetic milieu, including the renin-angiotensin system and transforming growth factor-β. The nature of the upstream modulators of Nox enzymes as well as the downstream targets of the Nox NADPH oxidases implicated in the propagation of the redox processes that alter renal biology in diabetes will be highlighted.
Collapse
Affiliation(s)
- Yves Gorin
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas,
USA
| | - Fabien Wauquier
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas,
USA
| |
Collapse
|
98
|
Kleino I, Järviluoma A, Hepojoki J, Huovila AP, Saksela K. Preferred SH3 domain partners of ADAM metalloproteases include shared and ADAM-specific SH3 interactions. PLoS One 2015; 10:e0121301. [PMID: 25825872 PMCID: PMC4380453 DOI: 10.1371/journal.pone.0121301] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 01/30/2015] [Indexed: 02/02/2023] Open
Abstract
A disintegrin and metalloproteinases (ADAMs) constitute a protein family essential for extracellular signaling and regulation of cell adhesion. Catalytic activity of ADAMs and their predicted potential for Src-homology 3 (SH3) domain binding show a strong correlation. Here we present a comprehensive characterization of SH3 binding capacity and preferences of the catalytically active ADAMs 8, 9, 10, 12, 15, 17, and 19. Our results revealed several novel interactions, and also confirmed many previously reported ones. Many of the identified SH3 interaction partners were shared by several ADAMs, whereas some were ADAM-specific. Most of the ADAM-interacting SH3 proteins were adapter proteins or kinases, typically associated with sorting and endocytosis. Novel SH3 interactions revealed in this study include TOCA1 and CIP4 as preferred partners of ADAM8, and RIMBP1 as a partner of ADAM19. Our results suggest that common as well as distinct mechanisms are involved in regulation and execution of ADAM signaling, and provide a useful framework for addressing the pathways that connect ADAMs to normal and aberrant cell behavior.
Collapse
Affiliation(s)
- Iivari Kleino
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Annika Järviluoma
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ari Pekka Huovila
- Institute of Biosciences and Medical Technology, University of Tampere, Tampere, Finland
| | - Kalle Saksela
- Department of Virology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- * E-mail:
| |
Collapse
|
99
|
|
100
|
Artym VV, Swatkoski S, Matsumoto K, Campbell CB, Petrie RJ, Dimitriadis EK, Li X, Mueller SC, Bugge TH, Gucek M, Yamada KM. Dense fibrillar collagen is a potent inducer of invadopodia via a specific signaling network. ACTA ACUST UNITED AC 2015; 208:331-50. [PMID: 25646088 PMCID: PMC4315243 DOI: 10.1083/jcb.201405099] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
High-density fibrillar collagen matrix induces invadopodia formation in both fibroblasts and carcinoma cell lines through a kindlin2-dependent mechanism that drives local ECM remodeling. Cell interactions with the extracellular matrix (ECM) can regulate multiple cellular activities and the matrix itself in dynamic, bidirectional processes. One such process is local proteolytic modification of the ECM. Invadopodia of tumor cells are actin-rich proteolytic protrusions that locally degrade matrix molecules and mediate invasion. We report that a novel high-density fibrillar collagen (HDFC) matrix is a potent inducer of invadopodia, both in carcinoma cell lines and in primary human fibroblasts. In carcinoma cells, HDFC matrix induced formation of invadopodia via a specific integrin signaling pathway that did not require growth factors or even altered gene and protein expression. In contrast, phosphoproteomics identified major changes in a complex phosphosignaling network with kindlin2 serine phosphorylation as a key regulatory element. This kindlin2-dependent signal transduction network was required for efficient induction of invadopodia on dense fibrillar collagen and for local degradation of collagen. This novel phosphosignaling mechanism regulates cell surface invadopodia via kindlin2 for local proteolytic remodeling of the ECM.
Collapse
Affiliation(s)
- Vira V Artym
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892 Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School; and Department of Biostatistics, Bioinformatics, and Biomathematics; Georgetown University, Washington, DC 20057
| | - Stephen Swatkoski
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Kazue Matsumoto
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Catherine B Campbell
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Ryan J Petrie
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Emilios K Dimitriadis
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Xin Li
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School; and Department of Biostatistics, Bioinformatics, and Biomathematics; Georgetown University, Washington, DC 20057
| | - Susette C Mueller
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School; and Department of Biostatistics, Bioinformatics, and Biomathematics; Georgetown University, Washington, DC 20057
| | - Thomas H Bugge
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Marjan Gucek
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| | - Kenneth M Yamada
- Laboratory of Cell and Developmental Biology, National Institute of Dental and Craniofacial Research; Proteomics Core Facility, National Heart, Lung, and Blood Institute; Biomolecular Engineering and Physical Sciences Shared Resource Program, National Institute of Biomolecular Imaging and Bioengineering; Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research; National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|