1
|
Johnson HE, Umutesi HG, Heo J. The small GTPase Rap1A expedites the NOX2 oxidative burst by facilitating Rac and NOX2 autoactivations. FEBS J 2025. [PMID: 40259664 DOI: 10.1111/febs.70107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/21/2025] [Accepted: 04/11/2025] [Indexed: 04/23/2025]
Abstract
Rac and Rap1A are small GTPases with the redox-sensitive GX4GK(S/T)C/ECS and NKCD motif. Of the known NADPH oxidase (NOX) isoforms, NOX1 and NOX2 function with the redox-sensitive Rac. Both exhibit an oxidative burst in which superoxide production is initially lagged but then accelerated. This burst is a reflection of NOX1 and NOX2 autoactivations occurring alongside the redox-dependent Rac autoactivation. NOX2 also contains the redox-sensitive Rap1A. However, its role in NOX2 function was unknown. In this study, we show that Rap1A is also autoactivated by its redox response, which is coupled to Rac and NOX2 autoactivations. This coupling is found to be mediated through the Rap1A-dependent recruitment of the Rac GEF P-REX1 to the NOX2 system. We further show that the initiation threshold and propagation rate of Rap1A autoactivation are lower and slower, respectively, than those of Rac and NOX2. The low-threshold Rap1A autoactivation recruits P-REX1 to the NOX2 system, resulting in the production of active Rac, thereby aiding the high-threshold initiation and propagation of Rac and NOX2 autoactivations. This results in the rapid completion of the NOX2 oxidative burst, which is specific to NOX2 because NOX1 lacks Rap1A. The redox response differences between the Rap1A NKCD motif and the Rac GX4GK(S/T)C/ECS motif appear to be the basis for the feature differences between Rap1A autoactivation and those of Rac and NOX2 autoactivations. The GX4GK(S/T)C/ECS and NKCD motifs are found in many redox-sensitive Rho/Rab and Ras family GTPases, respectively. Findings here shed light on previously unknown redox-dependent functional distinctions between these small GTPases.
Collapse
Affiliation(s)
- Hope Elizabeth Johnson
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX, USA
| | - Hope Gloria Umutesi
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX, USA
| | - Jongyun Heo
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
2
|
Engelhardt D, Petersen JR, Martyr C, Kuhn-Gale H, Niswander LA. Moderate levels of folic acid benefit outcomes for cilia based neural tube defects. Dev Biol 2025; 520:62-74. [PMID: 39755226 DOI: 10.1016/j.ydbio.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/20/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025]
Abstract
Folic acid (FA) supplementation is a potent tool to reduce devastating birth defects known as neural tube defects (NTDs). Though effective, questions remain how FA achieves its protective effect and which gene mutations are sensitive to folic acid levels. We explore the relationship between FA dosage and NTD rates using NTD mouse models. We demonstrate that NTD rates in mouse models harboring mutations in cilia genes depend on FA dosage. Cilia mutant mouse models demonstrate reductions in NTD rates when exposed to moderate levels of FA that are not observed at higher fortified levels of FA. This trend continues with a moderate level of FA being beneficial for primary and motile cilia formation. We present a mechanism through which fortified FA levels reduce basal levels of reactive oxygen species (ROS) which in turn reduces ROS-sensitive GTPase activity required for ciliogenesis. Our data indicates that genes involved in cilia formation and function represent a FA sensitive category of mutations and a possible avenue for further reducing NTD and ciliopathy incidences.
Collapse
Affiliation(s)
- David Engelhardt
- Department of Molecular, Cellular and Development Biology, University of Colorado, Boulder, CO, 80309, USA
| | - Juliette R Petersen
- Molecular Biology Graduate Program, University of Colorado Anschutz Medical Campus, Denver, CO, 80045, USA
| | - Cara Martyr
- Department of Molecular, Cellular and Development Biology, University of Colorado, Boulder, CO, 80309, USA
| | - Hannah Kuhn-Gale
- Department of Molecular, Cellular and Development Biology, University of Colorado, Boulder, CO, 80309, USA
| | - Lee A Niswander
- Department of Molecular, Cellular and Development Biology, University of Colorado, Boulder, CO, 80309, USA.
| |
Collapse
|
3
|
Salamh S, Sayyed-Ahmad A. Investigating the effects of cysteine-118 oxidation on G12D KRas structure and dynamics: insights from MD simulations. J Biomol Struct Dyn 2024; 42:6968-6981. [PMID: 37480262 DOI: 10.1080/07391102.2023.2238080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
Mutations of Ras proteins are believed to be among the most prominent causes of cancer. There is increasing evidence that the activity of Ras may be controlled by the redox state of cysteine residues located within the NKCD motif. This redox signaling is critical to both physiological and pathological processes and occurs when C118 is oxidized in a reversible manner. In this study, we used atomistic molecular dynamics simulations and Markov state models to investigate the structural and conformational effects of C118 oxidation on the oncogenic mutant KRas(G12D). While both mutants share common features and exhibit some distinct conformational states and fluctuations, we have found that the oxidized variant KRas(G12D/C118SOH) is more dynamic than the unoxidized counterpart, particularly in the switch II region. Additionally, C118 oxidation is found to alter the structure of the nucleotide-binding site and the switch regions as well as perturb the conformational equilibrium between Ras active and inactive states. These conformational preferences may alter the affinity to different effectors, resulting in selective downstream activation. Our results are anticipated to help future drug development efforts aimed at KRAS-related anticancer treatment.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shimaa Salamh
- Department of Physics, Birzeit University, Birzeit, Palestine
| | | |
Collapse
|
4
|
Zuchegna C, Porcellini A, Messina S. Redox-sensitive small GTPase H-Ras in murine astrocytes, an in vitro study. Redox Rep 2022; 27:150-157. [PMID: 35822835 PMCID: PMC9291712 DOI: 10.1080/13510002.2022.2094109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Although the protooncogenes small GTPases Ras are redox-sensitive proteins, how they are regulated by redox signaling in the central nervous system (CNS) is still poorly understood. Alteration in redox-sensitive targets by redox signaling may have myriad effects on Ras stability, activity and localization. Redox-mediated changes in astrocytic RAS may contribute to the control of redox homeostasis in the CNS that is connected to the pathogenesis of many diseases. RESULTS AND METHODS Here, we investigated the transient physiological induction, at both transcriptional and translational levels, of small GTPases Ras in response to redox stimulation. Cultured astrocytes were treated with hydrogen peroxide as in bolus addition and relative mRNA levels of murine hras and kras genes were detected by qRT-PCR. We found that de novo transcription of hras mRNA in reactive astrocytes is redox-sensitive and mimics the prototypical redox-sensitive gene iNOS. Protein abundance in combination with protein turnover measurements by cycloheximide-chase experiments revealed distinct translation efficiency, GTP-bound enrichment, and protein turnover rates between the two isoforms H-Ras and K-Ras. CONCLUSION Reports from recent years support a significant role of H-Ras in driving redox processes. Beyond its canonical functions, Ras may impact on the core astrocytic cellular machinery that operates during redox stimulation.
Collapse
Affiliation(s)
- Candida Zuchegna
- Dipartimento di Biologia, Complesso Universitario di Monte Sant'Angelo, Università degli Studi di Napoli “Federico II”, Napoli, Italia
| | - Antonio Porcellini
- Dipartimento di Biologia, Complesso Universitario di Monte Sant'Angelo, Università degli Studi di Napoli “Federico II”, Napoli, Italia
| | - Samantha Messina
- Dipartimento di Scienze, Università degli Studi Roma Tre, Roma, Italia
| |
Collapse
|
5
|
Medvedeva AV, Rebrova AV, Zalomaeva ES, Turaeva SK, Nikitina EA, Tokmacheva EV, Vasileva SA, Shchegolev BF, Savvateeva-Popova EV. Role of LIM Kinase 1 in Dopaminergic and Serotonergic Neurons in Genome Stability, Learning and Memory during Stress Response to Weakening of Earth’s Magnetic Field in Drosophila. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022010033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
6
|
Patel A, Perl A. Redox Control of Integrin-Mediated Hepatic Inflammation in Systemic Autoimmunity. Antioxid Redox Signal 2022; 36:367-388. [PMID: 34036799 PMCID: PMC8982133 DOI: 10.1089/ars.2021.0068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 12/20/2022]
Abstract
Significance: Systemic autoimmunity affects 3%-5% of the population worldwide. Systemic lupus erythematosus (SLE) is a prototypical form of such condition, which affects 20-150 of 100,000 people globally. Liver dysfunction, defined by increased immune cell infiltration into the hepatic parenchyma, is an understudied manifestation that affects up to 20% of SLE patients. Autoimmunity in SLE involves proinflammatory lineage specification in the immune system that occurs with oxidative stress and profound changes in cellular metabolism. As the primary metabolic organ of the body, the liver is uniquely capable to encounter oxidative stress through first-pass derivatization and filtering of waste products. Recent Advances: The traffic of immune cells from their development through recirculation in the liver is guided by cell adhesion molecules (CAMs) and integrins, cell surface proteins that tightly anchor cells together. The surface expression of CAMs and integrins is regulated via endocytic traffic that is sensitive to oxidative stress. Reactive oxygen species (ROS) that elicit oxidative stress in the liver may originate from the mitochondria, the cytosol, or the cell membrane. Critical Issues: While hepatic ROS production is a source of vulnerability, it also modulates the development and function of the immune system. In turn, the liver employs antioxidant defense mechanisms to protect itself from damage that can be harnessed to serve as therapeutic mechanisms against autoimmunity, inflammation, and development of hepatocellular carcinoma. Future Directions: This review is aimed at delineating redox control of integrin signaling in the liver and checkpoints of regulatory impact that can be targeted for treatment of inflammation in systemic autoimmunity. Antioxid. Redox Signal. 36, 367-388.
Collapse
Affiliation(s)
- Akshay Patel
- Division of Rheumatology, Department of Medicine, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- Department of Microbiology and Immunology, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Andras Perl
- Division of Rheumatology, Department of Medicine, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- Department of Microbiology and Immunology, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, State University of New York Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
7
|
Hurst M, McGarry DJ, Olson MF. Rho GTPases: Non-canonical regulation by cysteine oxidation. Bioessays 2021; 44:e2100152. [PMID: 34889471 DOI: 10.1002/bies.202100152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/20/2022]
Abstract
Rho GTPases are critically important and are centrally positioned regulators of the actomyosin cytoskeleton. By influencing the organization and architecture of the cytoskeleton, Rho proteins play prominent roles in many cellular processes including adhesion, migration, intra-cellular transportation, and proliferation. The most important method of Rho GTPase regulation is via the GTPase cycle; however, post-translational modifications (PTMs) also play critical roles in Rho protein regulation. Relative to other PTMs such as lipidation or phosphorylation that have been extensively characterized, protein oxidation is a regulatory PTM that has been poorly studied. Protein oxidation primarily occurs from the reaction of reactive oxygen species (ROS), such as hydrogen peroxide (H2 O2 ), with amino acid side chain thiols on cysteine (Cys) and methionine (Met) residues. The versatile redox modifications of cysteine residues exemplify their integral role in cell signalling processes. Here we review prominent members of the Rho GTPase family and discuss how lipidation, phosphorylation, and oxidation on conserved cysteine residues affects their regulation and function.
Collapse
Affiliation(s)
- Mackenzie Hurst
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - David J McGarry
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| | - Michael F Olson
- Department of Chemistry and Biology, Ryerson University, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Peptide DR8 analogs alleviate pulmonary fibrosis via suppressing TGF-β1 mediated epithelial-mesenchymal transition and ERK1/2 pathway in vivo and in vitro. Eur J Pharm Sci 2021; 167:106009. [PMID: 34537373 DOI: 10.1016/j.ejps.2021.106009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/26/2021] [Accepted: 09/14/2021] [Indexed: 02/06/2023]
Abstract
Pulmonary fibrosis is a chronic progressive lung disease that lacks effective treatments in clinic. It is characterized by repair disorder of epithelial cells, formation of fibroblast foci as well as destruction of alveolar structure. Previously we first determined that parent peptide DR8 (DHNNPQIR-NH2) has anti-fibrotic activity in bleomycin-induced mice. In order to further improve the druggability of DR8, including anti-fibrotic activity, stability and security, the structure-activity relationship was investigated using a series of D-amino acid and alanine scanning analogs of DR8. The results indicated that peptides DR8-3D and DR8-8A exhibited potent anti-fibrotic activity and better stability. Further mechanism research revealed that DR8-3D and DR8-8A ameliorated lung fibrosis by inhibiting TGF-β1 mediated epithelial-mesenchymal transition process and ERK1/2 signaling pathway in vitro and in vivo. Moreover, we found that anti-fibrotic activity of DR8 was closely related to the residues aspartic acid (Asp)1, histidine (His)2, proline (Pro)5 and glutamine (Gln)6, which suggested that the position of residues asparagine (Asn)3, asparagine (Asn)4, isoleucine (Ile)7 and arginine (Arg)8 could be further modified to optimized its anti-fibrotic effect. Therefore, we consider that DR8-3D and DR8-8A not only could be used as a potential leading compound for the treatment of bleomycin-induced lung fibrosis but also laid a foundation for the development of new anti-fibrotic drugs.
Collapse
|
9
|
Post-Translational Modification and Subcellular Compartmentalization: Emerging Concepts on the Regulation and Physiopathological Relevance of RhoGTPases. Cells 2021; 10:cells10081990. [PMID: 34440759 PMCID: PMC8393718 DOI: 10.3390/cells10081990] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/26/2022] Open
Abstract
Cells and tissues are continuously exposed to both chemical and physical stimuli and dynamically adapt and respond to this variety of external cues to ensure cellular homeostasis, regulated development and tissue-specific differentiation. Alterations of these pathways promote disease progression-a prominent example being cancer. Rho GTPases are key regulators of the remodeling of cytoskeleton and cell membranes and their coordination and integration with different biological processes, including cell polarization and motility, as well as other signaling networks such as growth signaling and proliferation. Apart from the control of GTP-GDP cycling, Rho GTPase activity is spatially and temporally regulated by post-translation modifications (PTMs) and their assembly onto specific protein complexes, which determine their controlled activity at distinct cellular compartments. Although Rho GTPases were traditionally conceived as targeted from the cytosol to the plasma membrane to exert their activity, recent research demonstrates that active pools of different Rho GTPases also localize to endomembranes and the nucleus. In this review, we discuss how PTM-driven modulation of Rho GTPases provides a versatile mechanism for their compartmentalization and functional regulation. Understanding how the subcellular sorting of active small GTPase pools occurs and what its functional significance is could reveal novel therapeutic opportunities.
Collapse
|
10
|
Rac-dependent feedforward autoactivation of NOX2 leads to oxidative burst. J Biol Chem 2021; 297:100982. [PMID: 34293347 PMCID: PMC8353492 DOI: 10.1016/j.jbc.2021.100982] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/09/2021] [Accepted: 07/17/2021] [Indexed: 12/03/2022] Open
Abstract
NADPH oxidase 2 (NOX2) produces the superoxide anion radical (O2−), which has functions in both cell signaling and immune defense. NOX2 is a multimeric-protein complex consisting of several protein subunits including the GTPase Rac. NOX2 uniquely facilitates an oxidative burst, which is described by initially slow O2− production, which increases over time. The NOX2 oxidative burst is considered critical to immune defense because it enables expedited O2− production in response to infections. However, the mechanism of the initiation and progression of this oxidative burst and its implications for regulation of NOX2 have not been clarified. In this study, we show that the NOX2 oxidative burst is a result of autoactivation of NOX2 coupled with the redox function of Rac. NOX2 autoactivation begins when active Rac triggers NOX2 activation and the subsequent production of O2−, which in turn activates redox-sensitive Rac. This activated Rac further activates NOX2, amplifying the feedforward cycle and resulting in a NOX2-mediated oxidative burst. Using mutagenesis-based kinetic and cell analyses, we show that enzymatic activation of Rac is exclusively responsible for production of the active Rac trigger that initiates NOX2 autoactivation, whereas redox-mediated Rac activation is the main driving force of NOX2 autoactivation and contributes to generation of ∼98% of the active NOX2 in cells. The results of this study provide insight into the regulation of NOX2 function, which could be used to develop therapeutics to control immune responses associated with dysregulated NOX2 oxidative bursts.
Collapse
|
11
|
Cuervo W, Sordillo LM, Abuelo A. Oxidative Stress Compromises Lymphocyte Function in Neonatal Dairy Calves. Antioxidants (Basel) 2021; 10:antiox10020255. [PMID: 33562350 PMCID: PMC7915147 DOI: 10.3390/antiox10020255] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 12/02/2022] Open
Abstract
Dairy calves are unable to mount an effective immune response during their first weeks of life, which contributes to increased disease susceptibility during this period. Oxidative stress (OS) diminishes the immune cell capabilities of humans and adult cows, and dairy calves also experience OS during their first month of life. However, the impact that OS may have on neonatal calf immunity remains unexplored. Thus, we aimed to evaluate the impact of OS on newborn calf lymphocyte functions. For this, we conducted two experiments. First, we assessed the association of OS status throughout the first month of age and the circulating concentrations of the cytokines interferon-gamma (IFN-γ) and interleukin (IL) 4, as well as the expression of cytokine-encoding genes IFNG, IL2, IL4, and IL10 in peripheral mononuclear blood cells (PBMCs) of 12 calves. Subsequently, we isolated PBMCs from another 6 neonatal calves to investigate in vitro the effect of OS on immune responses in terms of activation of lymphocytes, cytokine expression, and antibody production following stimulation with phorbol 12-myristate 13-acetate or bovine herpesvirus-1. The results were compared statistically through mixed models. Calves exposed to high OS status in their first month of age showed higher concentrations of IL-4 and expression of IL4 and IL10 and lower concentrations of IFN-γ and expression of IFNG and IL2 than calves exposed to lower OS. In vitro, OS reduced lymphocyte activation, production of antibodies, and protein and gene expression of key cytokines. Collectively, our results demonstrate that OS can compromise some immune responses of newborn calves. Hence, further studies are needed to explore the mechanisms of how OS affects the different lymphocyte subsets and the potential of ameliorating OS in newborn calves as a strategy to augment the functional capacity of calf immune cells, as well as enhance calves’ resistance to infections.
Collapse
|
12
|
Ngo ATP, Parra-Izquierdo I, Aslan JE, McCarty OJT. Rho GTPase regulation of reactive oxygen species generation and signalling in platelet function and disease. Small GTPases 2021; 12:440-457. [PMID: 33459160 DOI: 10.1080/21541248.2021.1878001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Platelets are master regulators and effectors of haemostasis with increasingly recognized functions as mediators of inflammation and immune responses. The Rho family of GTPase members Rac1, Cdc42 and RhoA are known to be major components of the intracellular signalling network critical to platelet shape change and morphological dynamics, thus playing a major role in platelet spreading, secretion and thrombus formation. Initially linked to the regulation of actomyosin contraction and lamellipodia formation, recent reports have uncovered non-canonical functions of platelet RhoGTPases in the regulation of reactive oxygen species (ROS), where intrinsically generated ROS modulate platelet function and contribute to thrombus formation. Platelet RhoGTPases orchestrate oxidative processes and cytoskeletal rearrangement in an interconnected manner to regulate intracellular signalling networks underlying platelet activity and thrombus formation. Herein we review our current knowledge of the regulation of platelet ROS generation by RhoGTPases and their relationship with platelet cytoskeletal reorganization, activation and function.
Collapse
Affiliation(s)
- Anh T P Ngo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| | - Ivan Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph E Aslan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA.,Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
13
|
Kramer-Drauberg M, Ambrogio C. Discoveries in the redox regulation of KRAS. Int J Biochem Cell Biol 2020; 131:105901. [PMID: 33309959 DOI: 10.1016/j.biocel.2020.105901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/27/2020] [Accepted: 12/05/2020] [Indexed: 10/22/2022]
Abstract
Oncogenic KRAS is one of the most common drivers of human cancer. Despite intense research, no effective therapy to directly inhibit oncogenic KRAS has yet been approved and KRAS mutant tumors remain associated with a poor prognosis. This short review discusses the current knowledge of the redox regulation of RAS and examines the newest findings on cysteine 118 (C118) as a potential novel target for KRAS inhibition.
Collapse
Affiliation(s)
- Maximilian Kramer-Drauberg
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
14
|
Lu WC, Xie H, Yuan C, Li JJ, Li ZY, Wu AH. Genomic landscape of the immune microenvironments of brain metastases in breast cancer. J Transl Med 2020; 18:327. [PMID: 32867782 PMCID: PMC7461335 DOI: 10.1186/s12967-020-02503-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 08/26/2020] [Indexed: 01/19/2023] Open
Abstract
Background This study was intended to investigate the genomic landscape of the immune microenvironments of brain metastases in breast cancer. Methods Three gene expression profile datasets (GSE76714, GSE125989 and GSE43837) of breast cancer with brain metastases were downloaded from Gene Expression Omnibus (GEO) database. After differential expression analysis, the tumor immune microenvironment and immune cell infiltration were analyzed. Then immune-related genes were identified, followed by function analysis, transcription factor (TF)-miRNA–mRNA co-regulatory network analysis, and survival analysis of metastatic recurrence. Results The present results showed that the tumor immune microenvironment in brain metastases was immunosuppressed compared with primary caner. Compared with primary cancer samples, the infiltration ratio of plasma cells in brain metastases samples was significantly higher, while the infiltration ratio of macrophages M2 cells in brain metastases samples was significantly lower. Total 42 immune-related genes were identified, such as THY1 and NEU2. CD1B, THY1 and DOCK2 were found to be implicated in the metastatic recurrence of breast cancer. Conclusions Targeting macrophages or plasma cells may be new strategies for immunotherapy of breast cancer with brain metastases. THY1 and NEU2 may be potential therapeutic targets for breast cancer with brain metastases, and THY1, CD1B and DOCK2 may serve as potential prognostic markers for improvement of brain metastases survival.
Collapse
Affiliation(s)
- Wei-Cheng Lu
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Hui Xie
- Department of Histology and Embryology, College of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning, China
| | - Ce Yuan
- Graduate Program in Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, USA
| | - Jin-Jiang Li
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Zhao-Yang Li
- Department of Laboratory Animal Center, China Medical University, Shenyang, Liaoning, China
| | - An-Hua Wu
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
15
|
Goloshvili G, Barbakadze T, Mikeladze D. Sodium nitroprusside induces H‐Ras depalmitoylation and alters the cellular response to hypoxia in differentiated and undifferentiated PC12 cells. Cell Biochem Funct 2019; 37:545-552. [DOI: 10.1002/cbf.3431] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 06/27/2019] [Accepted: 08/05/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Galina Goloshvili
- Faculty of Natural Sciences and MedicineIlia State University Tbilisi Georgia
| | - Tamar Barbakadze
- Faculty of Natural Sciences and MedicineIlia State University Tbilisi Georgia
- Department of BiochemistryI. Beritashvili Center of Experimental Biomedicine Tbilisi Georgia
| | - David Mikeladze
- Faculty of Natural Sciences and MedicineIlia State University Tbilisi Georgia
- Department of BiochemistryI. Beritashvili Center of Experimental Biomedicine Tbilisi Georgia
| |
Collapse
|
16
|
Messina S, De Simone G, Ascenzi P. Cysteine-based regulation of redox-sensitive Ras small GTPases. Redox Biol 2019; 26:101282. [PMID: 31386964 PMCID: PMC6695279 DOI: 10.1016/j.redox.2019.101282] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/21/2019] [Accepted: 07/24/2019] [Indexed: 12/22/2022] Open
Abstract
Reactive oxygen and nitrogen species (ROS and RNS, respectively) activate the redox-sensitive Ras small GTPases. The three canonical genes (HRAS, NRAS, and KRAS) are archetypes of the superfamily of small GTPases and are the most common oncogenes in human cancer. Oncogenic Ras is intimately linked to redox biology, mainly in the context of tumorigenesis. The Ras protein structure is highly conserved, especially in effector-binding regions. Ras small GTPases are redox-sensitive proteins thanks to the presence of the NKCD motif (Asn116-Lys 117-Cys118-Asp119). Notably, the ROS- and RNS-based oxidation of Cys118 affects protein stability, activity, and localization, and protein-protein interactions. Cys residues at positions 80, 181, 184, and 186 may also help modulate these actions. Moreover, oncogenic mutations of Gly12Cys and Gly13Cys may introduce additional oxidative centres and represent actionable drug targets. Here, the pathophysiological involvement of Cys-redox regulation of Ras proteins is reviewed in the context of cancer and heart and brain diseases.
Collapse
Affiliation(s)
- Samantha Messina
- Department of Science, Roma Tre University, Viale Guglielmo Marconi 446, I-00146, Roma, Italy.
| | - Giovanna De Simone
- Department of Science, Roma Tre University, Viale Guglielmo Marconi 446, I-00146, Roma, Italy
| | - Paolo Ascenzi
- Department of Science, Roma Tre University, Viale Guglielmo Marconi 446, I-00146, Roma, Italy
| |
Collapse
|
17
|
Sun W, Dai L, Yu H, Puspita B, Zhao T, Li F, Tan JL, Lim YT, Chen MW, Sobota RM, Tenen DG, Prabhu N, Nordlund P. Monitoring structural modulation of redox-sensitive proteins in cells with MS-CETSA. Redox Biol 2019; 24:101168. [PMID: 30925293 PMCID: PMC6439307 DOI: 10.1016/j.redox.2019.101168] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/07/2019] [Accepted: 03/10/2019] [Indexed: 12/31/2022] Open
Abstract
Reactive oxygen species (ROS) induce different cellular stress responses but can also mediate cellular signaling. Augmented levels of ROS are associated with aging, cancer as well as various metabolic and neurological disorders. ROS can also affect the efficacy and adverse effects of drugs. Although proteins are key mediators of most ROS effects, direct studies of ROS-modulated-protein function in the cellular context are very challenging. Therefore the understanding of specific roles of different proteins in cellular ROS responses is still relatively rudimentary. In the present work we show that Mass Spectrometry-Cellular Thermal Shift Assay (MS-CETSA) can directly monitor ROS and redox modulations of protein structure at the proteome level. By altering ROS levels in cultured human hepatocellular carcinoma cell lysates and intact cells, we detected CETSA responses in many proteins known to be redox sensitive, and also revealed novel candidate ROS sensitive proteins. Studies in intact cells treated with hydrogen peroxide and sulfasalazine, a ROS modulating drug, identified not only proteins that are directly modified, but also proteins reporting on downstream cellular effects. Comprehensive changes are seen on rate-limiting proteins regulating key cellular processes, including known redox control systems, protein degradation, epigenetic control and protein translational processes. Interestingly, concerted shifts on ATP-binding proteins revealed redox-induced modulation of ATP levels, which likely control many cellular processes. Collectively, these studies establish CETSA as a novel method for cellular studies of redox modulations of proteins, which implicated in a wide range of processes and for the discovery of CETSA-based biomarkers reporting on the efficacy as well as adverse effects of drugs.
Collapse
Affiliation(s)
- Wendi Sun
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Lingyun Dai
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Han Yu
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Brenda Puspita
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Tianyun Zhao
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Feng Li
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Justin L Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Genome Institute of Singapore, A*STAR, 138672, Singapore
| | - Yan Ting Lim
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Ming Wei Chen
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | | | - Daniel G Tenen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nayana Prabhu
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Pär Nordlund
- School of Biological Sciences, Nanyang Technological University, 637551, Singapore; Institute of Molecular and Cell Biology, A*STAR, 138673, Singapore; Department of Oncology and Pathology, Karolinska Institutet, Stockholm, 17177, Sweden.
| |
Collapse
|
18
|
Foo CHJ, Pervaiz S. gRASping the redox lever to modulate cancer cell fate signaling. Redox Biol 2019; 25:101094. [PMID: 30638892 PMCID: PMC6859584 DOI: 10.1016/j.redox.2018.101094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/22/2018] [Accepted: 12/26/2018] [Indexed: 01/17/2023] Open
Abstract
RAS proteins are critical regulators of signaling networks controlling diverse cellular functions such as cell proliferation and survival and its mutation are among the most powerful oncogenic drivers in human cancers. Despite intense efforts, direct RAS-targeting strategies remain elusive due to its "undruggable" nature. To that end, bulk of the research efforts has been directed towards targeting upstream and/or downstream of RAS signaling. However, the therapeutic efficacies of these treatments are limited in the long run due to the acquired drug resistance in RAS-driven cancers. Interestingly, recent studies have uncovered a potential role of RAS in redox-regulation as well as the interplay between ROS and RAS-associated signaling networks during process of cancer initiation and progression. More specifically, these studies provide ample evidence to implicate RAS as a redox-rheostat, manipulating ROS levels to provide a redox-milieu conducive for carcinogenesis. Importantly, the understanding of RAS-ROS interplay could provide us with novel targetable vulnerabilities for designing therapeutic strategies. In this review, we provide a brief summary of the advances in the field to illustrate the dual role of RAS in redox-regulation and its implications in RAS signaling outcomes and also emerging redox-based strategies to target RAS-driven cancers.
Collapse
Affiliation(s)
- Chuan Han Jonathan Foo
- Department of Physiology, YLL School of Medicine, National University of Singapore (NUS), Singapore; NUS Graduate School of Integrative Sciences and Engineering, NUS, Singapore
| | - Shazib Pervaiz
- Department of Physiology, YLL School of Medicine, National University of Singapore (NUS), Singapore; Medical Science Cluster Cancer Program, YLL School of Medicine, National University of Singapore (NUS), Singapore; NUS Graduate School of Integrative Sciences and Engineering, NUS, Singapore; National University Cancer Institute, NUHS, Singapore.
| |
Collapse
|
19
|
Castillo B, Kim SH, Sharief M, Sun T, Kim LW. SodC modulates ras and PKB signaling in Dictyostelium. Eur J Cell Biol 2016; 96:1-12. [PMID: 27919433 DOI: 10.1016/j.ejcb.2016.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 11/16/2016] [Indexed: 01/12/2023] Open
Abstract
We have previously reported that the basal RasG activity is aberrantly high in cells lacking Superoxide dismutase C (SodC). Here we report that other Ras proteins such as RasC and RasD activities are not affected in sodC- cells and mutagenesis studies showed that the presence of the Cys118 in the Ras proteins is essential for the superoxide-mediated activation of Ras proteins in Dictyostelium. In addition to the loss of SodC, lack of extracellular magnesium ions increased the level of intracellular superoxide and active RasG proteins. Aberrantly active Ras proteins in sodC- cells persistently localized at the plasma membrane, but those in wild type cells under magnesium deficient medium exhibited intracellular vesicular localization. Interestingly, the aberrantly activated Ras proteins in wild type cells were largely insulated from their normal downstream events such as Phosphatidylinositol-3,4,5-P3 (PIP3) accumulation, Protein Kinase B (PKB) activation, and PKBs substrates phosphorylation. Intriguingly, however, aberrantly activated Ras proteins in sodC- cells were still engaged in signaling to their downstream targets, and thus excessive PKBs substrates phosphorylation persisted. In summary, we suggest that SodC and RasG proteins are essential part of a novel inhibitory mechanism that discourages oxidatively stressed cells from chemotaxis and thus inhibits the delivery of potentially damaged genome to the next generation.
Collapse
Affiliation(s)
- Boris Castillo
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Seon-Hee Kim
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Mujataba Sharief
- Biochemistry PhD Program, Florida International University, Miami, FL 33199, USA
| | - Tong Sun
- Department of Biological Sciences, Florida International University, Miami, FL 33199, USA
| | - Lou W Kim
- Biochemistry PhD Program, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
20
|
Shin JY, Wey M, Umutesi HG, Sun X, Simecka J, Heo J. Thiopurine Prodrugs Mediate Immunosuppressive Effects by Interfering with Rac1 Protein Function. J Biol Chem 2016; 291:13699-714. [PMID: 27189938 DOI: 10.1074/jbc.m115.694422] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Indexed: 12/19/2022] Open
Abstract
6-Thiopurine (6-TP) prodrugs include 6-thioguanine and azathioprine. Both are widely used to treat autoimmune disorders and certain cancers. This study showed that a 6-thioguanosine triphosphate (6-TGTP), converted in T-cells from 6-TP, targets Rac1 to form a disulfide adduct between 6-TGTP and the redox-sensitive GXXXXGK(S/T)C motif of Rac1. This study also showed that, despite the conservation of the catalytic activity of RhoGAP (Rho-specific GAP) on the 6-TGTP-Rac1 adduct to produce the biologically inactive 6-thioguanosine diphosphate (6-TGDP)-Rac1 adduct, RhoGEF (Rho-specific GEF) cannot exchange the 6-TGDP adducted on Rac1 with free guanine nucleotide. The biologically inactive 6-TGDP-Rac1 adduct accumulates in cells because of the ongoing combined actions of RhoGEF and RhoGAP. Because other Rho GTPases, such as RhoA and Cdc42, also possess the GXXXXGK(S/T)C motif, the proposed mechanism for the inactivation of Rac1 also applies to RhoA and Cdc42. However, previous studies have shown that CD3/CD28-stimulated T-cells contain more activated Rac1 than other Rho GTPases such as RhoA and Cdc42. Accordingly, Rac1 is the main target of 6-TP in activated T-cells. This explains the T-cell-specific Rac1-targeting therapeutic action of 6-TP that suppresses the immune response. This proposed mechanism for the action of 6-TP on Rac1 performs a critical role in demonstrating the capability to design a Rac1-targeting chemotherapeutic agent(s) for autoimmune disorders. Nevertheless, the results also suggest that the targeting action of other Rho GTPases in other organ cells, such as RhoA in vascular cells, may be linked to cytotoxicities because RhoA plays a key role in vasculature functions.
Collapse
Affiliation(s)
- Jin-Young Shin
- From the Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019
| | - Michael Wey
- From the Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019
| | - Hope G Umutesi
- From the Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019
| | - Xiangle Sun
- the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107, and
| | - Jerry Simecka
- the Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, Texas 76107, and the Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas 76107
| | - Jongyun Heo
- From the Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019,
| |
Collapse
|
21
|
Bräutigam L, Pudelko L, Jemth AS, Gad H, Narwal M, Gustafsson R, Karsten S, Carreras Puigvert J, Homan E, Berndt C, Berglund UW, Stenmark P, Helleday T. Hypoxic Signaling and the Cellular Redox Tumor Environment Determine Sensitivity to MTH1 Inhibition. Cancer Res 2016; 76:2366-75. [PMID: 26862114 DOI: 10.1158/0008-5472.can-15-2380] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/29/2016] [Indexed: 11/16/2022]
Abstract
Cancer cells are commonly in a state of redox imbalance that drives their growth and survival. To compensate for oxidative stress induced by the tumor redox environment, cancer cells upregulate specific nononcogenic addiction enzymes, such as MTH1 (NUDT1), which detoxifies oxidized nucleotides. Here, we show that increasing oxidative stress in nonmalignant cells induced their sensitization to the effects of MTH1 inhibition, whereas decreasing oxidative pressure in cancer cells protected against inhibition. Furthermore, we purified zebrafish MTH1 and solved the crystal structure of MTH1 bound to its inhibitor, highlighting the zebrafish as a relevant tool to study MTH1 biology. Delivery of 8-oxo-dGTP and 2-OH-dATP to zebrafish embryos was highly toxic in the absence of MTH1 activity. Moreover, chemically or genetically mimicking activated hypoxia signaling in zebrafish revealed that pathologic upregulation of the HIF1α response, often observed in cancer and linked to poor prognosis, sensitized embryos to MTH1 inhibition. Using a transgenic zebrafish line, in which the cellular redox status can be monitored in vivo, we detected an increase in oxidative pressure upon activation of hypoxic signaling. Pretreatment with the antioxidant N-acetyl-L-cysteine protected embryos with activated hypoxia signaling against MTH1 inhibition, suggesting that the aberrant redox environment likely causes sensitization. In summary, MTH1 inhibition may offer a general approach to treat cancers characterized by deregulated hypoxia signaling or redox imbalance. Cancer Res; 76(8); 2366-75. ©2016 AACR.
Collapse
Affiliation(s)
- Lars Bräutigam
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| | - Linda Pudelko
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ann-Sofie Jemth
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Helge Gad
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Mohit Narwal
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Robert Gustafsson
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Stella Karsten
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jordi Carreras Puigvert
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Evert Homan
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Life Science Center, Düsseldorf, Germany
| | - Ulrika Warpman Berglund
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Thomas Helleday
- Science for Life Laboratory, Division of Translational Medicine and Chemical Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
22
|
Abstract
The genetic landscape of pancreatic cancer shows nearly ubiquitous mutations of K-RAS. However, oncogenic K-Rasmt alone is not sufficient to lead to pancreatic ductal adenocarcinoma (PDAC) in either human or in genetically modified adult mouse models. Many stimulants, such as high fat diet, CCK, LPS, PGE2 and others, have physiological effects at low concentrations that are mediated in part through modest increases in K-Ras activity. However, at high concentrations, they induce inflammation that, in the presence of oncogenic K-Ras expression, substantially accelerates PDAC formation. The mechanism involves increased activity of oncogenic K-Rasmt. Unlike what has been proposed in the standard paradigm for the role of Ras in oncogenesis, oncogenic K-Rasmt is now known to not be constitutively active. Rather, it can be activated by standard mechanisms similar to wild-type K-Ras, but its activity is sustained for a prolonged period. Furthermore, if the level of K-Ras activity exceeds a threshold at which it begins to generate its own activators, then a feed-forward loop is formed between K-Ras activity and inflammation and pathological processes including oncogenesis are initiated. Oncogenic K-Rasmt activation, a key event in PDAC initiation and development, is subject to complex regulatory mechanisms. Reagents which inhibit inflammation, such as the Cox2 inhibitor celecoxib, block the feed-forward loop and prevent induction of PDAC in models with endogenous oncogenic K-Rasmt. Increased understanding of the role of activating and inhibitory mechanisms on oncogenic K-Rasmt activity is of paramount importance for the development of preventive and therapeutic strategies to fight against this lethal disease.
Collapse
Affiliation(s)
- Craig D Logsdon
- 1. Department of GI Medical Oncology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA; 2. Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA
| | - Weiqin Lu
- 1. Department of GI Medical Oncology, University of Texas MD Anderson Cancer Center, Houston TX 77030, USA
| |
Collapse
|
23
|
Choudhary S, Boldogh I, Brasier AR. Inside-Out Signaling Pathways from Nuclear Reactive Oxygen Species Control Pulmonary Innate Immunity. J Innate Immun 2016; 8:143-55. [PMID: 26756522 PMCID: PMC4801701 DOI: 10.1159/000442254] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 02/05/2023] Open
Abstract
The airway mucosa is responsible for mounting a robust innate immune response (IIR) upon encountering pathogen-associated molecular patterns. The IIR produces protective gene networks that stimulate neighboring epithelia and components of the immune system to trigger adaptive immunity. Little is currently known about how cellular reactive oxygen species (ROS) signaling is produced and cooperates in the IIR. We discuss recent discoveries about 2 nuclear ROS signaling pathways controlling innate immunity. Nuclear ROS oxidize guanine bases to produce mutagenic 8-oxoguanine, a lesion excised by 8-oxoguanine DNA glycosylase1/AP-lyase (OGG1). OGG1 forms a complex with the excised base, inducing its nuclear export. The cytoplasmic OGG1:8-oxoG complex functions as a guanine nucleotide exchange factor, triggering small GTPase signaling and activating phosphorylation of the nuclear factor (NF)x03BA;B/RelA transcription factor to induce immediate early gene expression. In parallel, nuclear ROS are detected by ataxia telangiectasia mutated (ATM), a PI3 kinase activated by ROS, triggering its nuclear export. ATM forms a scaffold with ribosomal S6 kinases, inducing RelA phosphorylation and resulting in transcription-coupled synthesis of type I and type III interferons and CC and CXC chemokines. We propose that ATM and OGG1 are endogenous nuclear ROS sensors that transmit nuclear signals that coordinate with outside-in pattern recognition receptor signaling, regulating the IIR.
Collapse
Affiliation(s)
- Sanjeev Choudhary
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex., USA
- Department of Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex., USA
- Department of Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Tex., USA
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Tex., USA
- Department of Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex., USA
- Department of Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Tex., USA
| | - Allan R. Brasier
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Tex., USA
- Department of Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Tex., USA
- Department of Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Tex., USA
| |
Collapse
|
24
|
8-Oxoguanine DNA glycosylase-1-driven DNA base excision repair: role in asthma pathogenesis. Curr Opin Allergy Clin Immunol 2015; 15:89-97. [PMID: 25486379 DOI: 10.1097/aci.0000000000000135] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW To provide both an overview and evidence of the potential cause of oxidative DNA base damage and repair signaling in chronic inflammation and histological changes associated with asthma. RECENT FINDINGS Asthma is initiated/maintained by immunological, genetic/epigenetic, and environmental factors. It is a world-wide health problem, as current therapies suppress symptoms rather than prevent/reverse the disease, largely due to gaps in understanding its molecular mechanisms. Inflammation, oxidative stress, and DNA damage are inseparable phenomena, but their molecular roles in asthma pathogenesis are unclear. It was found that among oxidatively modified DNA bases, 8-oxoguanine (8-oxoG) is one of the most abundant, and its levels in DNA and body fluids are considered a biomarker of ongoing asthmatic processes. Free 8-oxoG forms a complex with 8-oxoG DNA glycosylase-1 and activates RAS-family GTPases that induce gene expression to mobilize innate and adaptive immune systems, along with genes regulating airway hyperplasia, hyper-responsiveness, and lung remodeling in atopic and nonatopic asthma. SUMMARY DNA's integrity must be maintained to prevent mutation, so its continuous repair and downstream signaling 'fuel' chronic inflammatory processes in asthma and form the basic mechanism whose elucidation will allow the development of new drug targets for the prevention/reversal of lung diseases.
Collapse
|
25
|
Aguilera-Aguirre L, Hosoki K, Bacsi A, Radák Z, Wood TG, Widen SG, Sur S, Ameredes BT, Saavedra-Molina A, Brasier AR, Ba X, Boldogh I. Whole transcriptome analysis reveals an 8-oxoguanine DNA glycosylase-1-driven DNA repair-dependent gene expression linked to essential biological processes. Free Radic Biol Med 2015; 81:107-18. [PMID: 25614460 PMCID: PMC4359954 DOI: 10.1016/j.freeradbiomed.2015.01.004] [Citation(s) in RCA: 31] [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: 11/01/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species inflict oxidative modifications on various biological molecules, including DNA. One of the most abundant DNA base lesions, 8-oxo-7,8-dihydroguanine (8-oxoG) is repaired by 8-oxoguanine DNA glycosylase-1 (OGG1) during DNA base excision repair (OGG1-BER). 8-OxoG accumulation in DNA has been associated with various pathological and aging processes, although its role is unclear. The lack of OGG1-BER in Ogg1(-/-) mice resulted in decreased inflammatory responses and increased susceptibility to infections and metabolic disorders. Therefore, we proposed that OGG1 and/or 8-oxoG base may have a role in immune and homeostatic processes. To test our hypothesis, we challenged mouse lungs with OGG1-BER product 8-oxoG base and changes in gene expression were determined by RNA sequencing and data were analyzed by Gene Ontology and statistical tools. RNA-Seq analysis identified 1592 differentially expressed (≥ 3-fold change) transcripts. The upregulated mRNAs were related to biological processes, including homeostatic, immune-system, macrophage activation, regulation of liquid-surface tension, and response to stimulus. These processes were mediated by chemokines, cytokines, gonadotropin-releasing hormone receptor, integrin, and interleukin signaling pathways. Taken together, these findings point to a new paradigm showing that OGG1-BER plays a role in various biological processes that may benefit the host, but when in excess could be implicated in disease and/or aging processes.
Collapse
Affiliation(s)
- Leopoldo Aguilera-Aguirre
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Koa Hosoki
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Attila Bacsi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Zsolt Radák
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thomas G Wood
- Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Steven G Widen
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Sanjiv Sur
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Bill T Ameredes
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alfredo Saavedra-Molina
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Xueqing Ba
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
26
|
Eller-Borges R, Batista WL, da Costa PE, Tokikawa R, Curcio MF, Strumillo ST, Sartori A, Moraes MS, de Oliveira GA, Taha MO, Fonseca FV, Stern A, Monteiro HP. Ras, Rac1, and phosphatidylinositol-3-kinase (PI3K) signaling in nitric oxide induced endothelial cell migration. Nitric Oxide 2015; 47:40-51. [PMID: 25819133 DOI: 10.1016/j.niox.2015.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 03/13/2015] [Accepted: 03/18/2015] [Indexed: 10/23/2022]
Abstract
The small GTP-binding proteins Ras and Rac1 are molecular switches exchanging GDP for GTP and converting external signals in response to a variety of stimuli. Ras and Rac1 play an important role in cell proliferation, cell differentiation, and cell migration. Rac1 is directly involved in the reorganization and changes in the cytoskeleton during cell motility. Nitric oxide (NO) stimulates the Ras - ERK1/2 MAP kinases signaling pathway and is involved in the interaction between Ras and the phosphatidyl-inositol-3 Kinase (PI3K) signaling pathway and cell migration. This study utilizes bradykinin (BK), which promotes endogenous production of NO, in an investigation of the role of NO in the activation of Rac1 in rabbit aortic endothelial cells (RAEC). NO-derived from BK stimulation of RAEC and incubation of the cells with the s-nitrosothiol S-nitrosoglutathione (GSNO) activated Rac1. NO-derived from BK stimulation promoted RAEC migration over a period of 12 h. The use of RAEC permanently transfected with the dominant negative mutant of Ras (Ras(N17)) or with the non-nitrosatable mutant of Ras (Ras(C118S)); and the use of specific inhibitors of: Ras, PI3K, and Rac1 resulted in inhibition of NO-mediated Rac1 activation. BK-stimulated s-nitrosylation of Ras in RAEC mediates Rac1 activation and cell migration. Inhibition of NO-mediated Rac1 activation resulted in inhibition of endothelial cell migration. In conclusion, the NO indirect activation of Rac1 involves the direct participation of Ras and PI3K in the migration of endothelial cells stimulated with BK.
Collapse
Affiliation(s)
- Roberta Eller-Borges
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Wagner L Batista
- Department of Biological Sciences, Universidade Federal de São Paulo/Campus Diadema, SP, Brazil
| | - Paulo E da Costa
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Rita Tokikawa
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Marli F Curcio
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Scheilla T Strumillo
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Adriano Sartori
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Miriam S Moraes
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Graciele A de Oliveira
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil
| | - Murched O Taha
- Department of Surgery, Escola Paulista de Medicina/Universidade Federal de São Paulo, SP, Brazil
| | - Fábio V Fonseca
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western University, Cleveland, OH, USA
| | - Arnold Stern
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA; Escuela de Medicina, Universidad Espíritu Santo, Guayaquil, Ecuador.
| | - Hugo P Monteiro
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMOL, Escola Paulista de Medicina /Universidade Federal de São Paulo, SP, Brazil.
| |
Collapse
|
27
|
Winterbourn CC. Are free radicals involved in thiol-based redox signaling? Free Radic Biol Med 2015; 80:164-70. [PMID: 25277419 DOI: 10.1016/j.freeradbiomed.2014.08.017] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Revised: 08/12/2014] [Accepted: 08/18/2014] [Indexed: 12/27/2022]
Abstract
Cells respond to many stimuli by transmitting signals through redox-regulated pathways. It is generally accepted that in many instances signal transduction is via reversible oxidation of thiol proteins, although there is uncertainty about the specific redox transformations involved. The prevailing view is that thiol oxidation occurs by a two electron mechanism, most commonly involving hydrogen peroxide. Free radicals, on the other hand, are considered as damaging species and not generally regarded as important in cell signaling. This paper examines whether it is justified to dismiss radicals or whether they could have a signaling role. Although there is no direct evidence that radicals are involved in transmitting thiol-based redox signals, evidence is presented that they are generated in cells when these signaling pathways are activated. Radicals produce the same thiol oxidation products as two electron oxidants, although by a different mechanism, and at this point radical-mediated pathways should not be dismissed. There are unresolved issues about how radical mechanisms could achieve sufficient selectivity, but this could be possible through colocalization of radical-generating and signal-transducing proteins. Colocalization is also likely to be important for nonradical signaling mechanisms and identification of such associations should be a priority for advancing the field.
Collapse
Affiliation(s)
- Christine C Winterbourn
- Centre for Free Radical Research, Department of Pathology, University of Otago, P.O. Box 4345, Christchurch, New Zealand.
| |
Collapse
|
28
|
Aguilera-Aguirre L, Bacsi A, Radak Z, Hazra TK, Mitra S, Sur S, Brasier AR, Ba X, Boldogh I. Innate inflammation induced by the 8-oxoguanine DNA glycosylase-1-KRAS-NF-κB pathway. THE JOURNAL OF IMMUNOLOGY 2014; 193:4643-53. [PMID: 25267977 DOI: 10.4049/jimmunol.1401625] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
8-Oxoguanine-DNA glycosylase-1 (OGG1) is the primary enzyme for repairing 7,8-dihydro-8-oxoguanine (8-oxoG) via the DNA base excision repair pathway (OGG1-BER). Accumulation of 8-oxoG in the genomic DNA leads to genetic instability and carcinogenesis and is thought to contribute to the worsening of various inflammatory and disease processes. However, the disease mechanism is unknown. In this study, we proposed that the mechanistic link between OGG1-BER and proinflammatory gene expression is OGG1's guanine nucleotide exchange factor activity, acquired after interaction with the 8-oxoG base and consequent activation of the small GTPase RAS. To test this hypothesis, we used BALB/c mice expressing or deficient in OGG1 in their airway epithelium and various molecular biological approaches, including active RAS pulldown, reporter and Comet assays, small interfering RNA-mediated depletion of gene expression, quantitative RT-PCR, and immunoblotting. We report that the OGG1-initiated repair of oxidatively damaged DNA is a prerequisite for GDP → GTP exchange, KRAS-GTP-driven signaling via MAP kinases and PI3 kinases and mitogen-stress-related kinase-1 for NF-κB activation, proinflammatory chemokine/cytokine expression, and inflammatory cell recruitment to the airways. Mice deficient in OGG1-BER showed significantly decreased immune responses, whereas a lack of other Nei-like DNA glycosylases (i.e., NEIL1 and NEIL2) had no significant effect. These data unveil a previously unidentified role of OGG1-driven DNA BER in the generation of endogenous signals for inflammation in the innate signaling pathway.
Collapse
Affiliation(s)
| | - Attila Bacsi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Zsolt Radak
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Tapas K Hazra
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555; Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555; and
| | - Sankar Mitra
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555
| | - Sanjiv Sur
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555; and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Allan R Brasier
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX 77555; and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555
| | - Xueqing Ba
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555; Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
29
|
The role of 8-oxoguanine DNA glycosylase-1 in inflammation. Int J Mol Sci 2014; 15:16975-97. [PMID: 25250913 PMCID: PMC4200771 DOI: 10.3390/ijms150916975] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/09/2014] [Accepted: 09/16/2014] [Indexed: 12/12/2022] Open
Abstract
Many, if not all, environmental pollutants/chemicals and infectious agents increase intracellular levels of reactive oxygen species (ROS) at the site of exposure. ROS not only function as intracellular signaling entities, but also induce damage to cellular molecules including DNA. Among the several dozen ROS-induced DNA base lesions generated in the genome, 8-oxo-7,8-dihydroguanine (8-oxoG) is one of the most abundant because of guanine’s lowest redox potential among DNA bases. In mammalian cells, 8-oxoG is repaired by the 8-oxoguanine DNA glycosylase-1 (OGG1)-initiated DNA base excision repair pathway (OGG1–BER). Accumulation of 8-oxoG in DNA has traditionally been associated with mutagenesis, as well as various human diseases and aging processes, while the free 8-oxoG base in body fluids is one of the best biomarkers of ongoing pathophysiological processes. In this review, we discuss the biological significance of the 8-oxoG base and particularly the role of OGG1–BER in the activation of small GTPases and changes in gene expression, including those that regulate pro-inflammatory chemokines/cytokines and cause inflammation.
Collapse
|
30
|
Abstract
SIGNIFICANCE We provide a conceptual framework for the interactions between the cellular redox signaling hub and the phytohormone signaling network that controls plant growth and development to maximize plant productivity under stress-free situations, while limiting growth and altering development on exposure to stress. RECENT ADVANCES Enhanced cellular oxidation plays a key role in the regulation of plant growth and stress responses. Oxidative signals or cycles of oxidation and reduction are crucial for the alleviation of dormancy and quiescence, activating the cell cycle and triggering genetic and epigenetic control that underpin growth and differentiation responses to changing environmental conditions. CRITICAL ISSUES The redox signaling hub interfaces directly with the phytohormone network in the synergistic control of growth and its modulation in response to environmental stress, but a few components have been identified. Accumulating evidence points to a complex interplay of phytohormone and redox controls that operate at multiple levels. For simplicity, we focus here on redox-dependent processes that control root growth and development and bud burst. FUTURE DIRECTIONS The multiple roles of reactive oxygen species in the control of plant growth and development have been identified, but increasing emphasis should now be placed on the functions of redox-regulated proteins, along with the central roles of reductants such as NAD(P)H, thioredoxins, glutathione, glutaredoxins, peroxiredoxins, ascorbate, and reduced ferredoxin in the regulation of the genetic and epigenetic factors that modulate the growth and vigor of crop plants, particularly within an agricultural context.
Collapse
Affiliation(s)
- Michael J Considine
- 1 School of Plant Biology and Institute of Agriculture, University of Western Australia , Crawley, Australia
| | | |
Collapse
|
31
|
Pandita TK. Unraveling the novel function of the DNA repair enzyme 8-oxoguanine-DNA glycosylase in activating key signaling pathways. Free Radic Biol Med 2014; 73:439-40. [PMID: 24878260 PMCID: PMC4151468 DOI: 10.1016/j.freeradbiomed.2014.05.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 05/19/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Tej K Pandita
- The Houston Methodist Research Institute, Houston, TX 77030, USA.
| |
Collapse
|
32
|
Wolff G, Balke JE, Andras IE, Park M, Toborek M. Exercise modulates redox-sensitive small GTPase activity in the brain microvasculature in a model of brain metastasis formation. PLoS One 2014; 9:e97033. [PMID: 24804765 PMCID: PMC4013134 DOI: 10.1371/journal.pone.0097033] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 04/14/2014] [Indexed: 12/26/2022] Open
Abstract
Tumor cell extravasation into the brain requires passage through the blood-brain barrier (BBB). There is evidence that exercise can alter the oxidation status of the brain microvasculature and protect against tumor cell invasion into the brain, although the mechanisms are not well understood. In the current study, we focused on the role of microenvironment generated by exercise and metastasizing tumor cells at the levels of brain microvessels, influencing oxidative stress-mediated responses and activation of redox-sensitive small GTPases. Mature male mice were exercised for four weeks using a running wheel with the average voluntary running distance 9.0 ± 0.3 km/day. Mice were then infused with 1.0 × 10(6) D122 (murine Lewis lung carcinoma) cells into the brain microvasculature, and euthanized either 48 hours (in short-term studies) or 2-3 weeks (in long-term studies) post tumor cell administration. A significant increase in the level of reactive oxygen species was observed following 48 hours or 3 weeks of tumor cells growth, which was accompanied by a reduction in MnSOD expression in the exercised mice. Activation of the small GTPase Rho was negatively correlated with running distance in the tumor cell infused mice. Together, these data suggest that exercise may play a significant role during aggressive metastatic invasion, especially at higher intensities in pre-trained individuals.
Collapse
Affiliation(s)
- Gretchen Wolff
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Jordan E. Balke
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Ibolya E. Andras
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Minseon Park
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, Florida, United States of America
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Miami, Florida, United States of America
- Jerzy Kukuczka Academy of Physical Education, Katowice, Poland
| |
Collapse
|
33
|
Stanley A, Thompson K, Hynes A, Brakebusch C, Quondamatteo F. NADPH oxidase complex-derived reactive oxygen species, the actin cytoskeleton, and Rho GTPases in cell migration. Antioxid Redox Signal 2014; 20:2026-42. [PMID: 24251358 DOI: 10.1089/ars.2013.5713] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
SIGNIFICANCE Rho GTPases are historically known to be central regulators of actin cytoskeleton reorganization. This affects many processes including cell migration. In addition, members of the Rac subfamily are known to be involved in reactive oxygen species (ROS) production through the regulation of NADPH oxidase (Nox) activity. This review focuses on relationships between Nox-regulated ROS, Rho GTPases, and cytoskeletal reorganization, in the context of cell migration. RECENT ADVANCES It has become clear that ROS participate in the regulation of certain Rho GTPase family members, thus mediating cytoskeletal reorganization. CRITICAL ISSUES The role of the actin cytoskeleton in providing a scaffold for components of the Nox complex needs to be examined in the light of these new advances. During cell migration, Rho GTPases, ROS, and cytoskeletal organization appear to function as a complex regulatory network. However, more work is needed to fully elucidate the interactions between these factors and their potential in vivo importance. FUTURE DIRECTIONS Ultrastructural analysis, that is, electron microscopy, particularly immunogold labeling, will enable direct visualization of subcellular compartments. This in conjunction with the analysis of tissues lacking specific Rho GTPases, and Nox components will facilitate a detailed examination of the interactions of these structures with the actin cytoskeleton. In combination with the analysis of ROS production, including its subcellular location, these data will contribute significantly to our understanding of this intricate network under physiological conditions. Based on this, in vivo and in vitro studies can then be combined to elucidate the signaling pathways involved and their targets.
Collapse
Affiliation(s)
- Alanna Stanley
- 1 Skin and Extracellular Matrix Research Group , Anatomy, NUI Galway, Galway, Ireland
| | | | | | | | | |
Collapse
|
34
|
T cell activation induces CuZn superoxide dismutase (SOD)-1 intracellular re-localization, production and secretion. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:265-74. [DOI: 10.1016/j.bbamcr.2013.10.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 10/21/2013] [Accepted: 10/24/2013] [Indexed: 01/22/2023]
|
35
|
Wey M, Phan V, Yepez G, Heo J. Superoxide inhibits guanine nucleotide exchange factor (GEF) action on Ras, but not on Rho, through desensitization of Ras to GEF. Biochemistry 2014; 53:518-32. [PMID: 24422478 PMCID: PMC4327825 DOI: 10.1021/bi401528n] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ras and Rho GTPases are molecular switches for various vital cellular signaling pathways. Overactivation of these GTPases often causes development of cancer. Guanine nucleotide exchange factors (GEFs) and oxidants function to upregulate these GTPases through facilitation of guanine nucleotide exchange (GNE) of these GTPases. However, the effect of oxidants on GEF functions, or vice versa, has not been known. We show that, via targeting Ras Cys(51), an oxidant inhibits the catalytic action of Cdc25-the catalytic domain of RasGEFs-on Ras. However, the enhancement of Ras GNE by an oxidant continues regardless of the presence of Cdc25. Limiting RasGEF action by an oxidant may function to prevent the pathophysiological overactivation of Ras in the presence of both RasGEFs and oxidants. The continuous exposure of Ras to nitric oxide and its derivatives can form S-nitrosated Ras (Ras-SNO). This study also shows that an oxidant not only inhibits the catalytic action of Cdc25 on Ras-SNO but also fails to enhance Ras-SNO GNE. This lack of enhancement then populates the biologically inactive Ras-SNO in cells, which may function to prevent the continued redox signaling of the Ras pathophysiological response. Finally, this study also demonstrates that, unlike the case with RasGEFs, an oxidant does not inhibit the catalytic action of RhoGEF-Vav or Dbs-on Rho GTPases such as Rac1, RhoA, RhoC, and Cdc42. This result explains the results of the previous study in which, despite the presence of an oxidant, the catalytic action of Dbs in cells continued to enhance RhoC GNE.
Collapse
Affiliation(s)
- Michael Wey
- Department of Chemistry and Biochemistry, The University of Texas at Arlington , Arlington, Texas 76019, United States
| | | | | | | |
Collapse
|
36
|
Abstract
Redox agents have been historically considered pathological agents which can react with and damage many biological macromolecules including DNA, proteins, and lipids. However, a growing number of reports have suggested that mammalian cells can rapidly respond to ligand stimulation with a change in intracellular ROS thus indicating that the production of intracellular redox agents is tightly regulated and that they serve as intracellular signaling molecules being involved in a variety of cell signaling pathways. Numerous observations have suggested that some members of the Ras GTPase superfamily appear to regulate the production of redox agents and that oxidants can function as effector molecules for the small GTPases, thus contributing to their overall biological function. In addition, many of the Ras superfamily small GTPases have been shown to be redox sensitive, thanks to the presence of redox-sensitive sequences in their primary structure. The action of redox agents on these redox-sensitive GTPases is similar to that of guanine nucleotide exchange factors in that they perturb GTPase nucleotide-binding interactions that result in the enhancement of the guanine nucleotide exchange of small GTPases. Thus, Ras GTPases may act both as upstream regulators and downstream effectors of redox agents. Here we overview current understanding concerning the interplay between Ras GTPases and redox agents, also taking into account pathological implications of misregulation of this cross talk and highlighting the potentiality of these cellular pathways as new therapeutical targets for different pathologies.
Collapse
|
37
|
Woolley JF, Corcoran A, Groeger G, Landry WD, Cotter TG. Redox-regulated growth factor survival signaling. Antioxid Redox Signal 2013. [PMID: 23198948 DOI: 10.1089/ars.2012.5028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Once the thought of as unwanted byproducts of cellular respiration in eukaryotes, reactive oxygen species (ROS) have been shown to facilitate essential physiological roles. It is now understood that ROS are critical mediators of intracellular signaling. Control of signal transduction downstream of growth factor receptors by ROS is a complex process whose details are only recently coming to light. RECENT ADVANCES Indeed, recent evidence points to control of signal propagation by ROS at multiple levels in the typical cascade. Growth factor stimulation activates nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Noxs) at the membrane, producing superoxide in the extracellular matrix, which is catalyzed to the membrane-permeable hydrogen peroxide (H2O2) that mediates intracellular signaling events. CRITICAL ISSUES The potential for H2O2, however, to disrupt cellular functions by damaging proteins and nucleic acids demands that its levels are kept in check by receptor-associated peroxiredoxins. This interplay of Nox and peroxiredoxin activity moderates levels of H2O2 sufficiently to modify signaling partners locally. Among the best studied of these partners are redox-controlled phosphatases that are inactivated by H2O2. Phosphatases regulate signal propagation downstream of receptors, and thus their inactivation allows a further level of control. Transmission of information further downstream to targets such as transcription factors, themselves regulated by ROS, completes this pathway. FUTURE DIRECTIONS Thus, signal propagation or attenuation can be dictated by ROS at multiple points. Given the complex nature of these processes, we envisage the emerging trends in the field of redox signaling in the context of growth factor stimulation.
Collapse
Affiliation(s)
- John F Woolley
- Tumour Biology Laboratory, Biochemistry Department, Bioscience Research Institute, University College , Cork, Ireland
| | | | | | | | | |
Collapse
|
38
|
Lu Y, Zhao X, Li K, Luo G, Nie Y, Shi Y, Zhou Y, Ren G, Feng B, Liu Z, Pan Y, Li T, Guo X, Wu K, Miranda-Vizuete A, Wang X, Fan D. Thioredoxin-like protein 2 is overexpressed in colon cancer and promotes cancer cell metastasis by interaction with ran. Antioxid Redox Signal 2013; 19:899-911. [PMID: 23311631 PMCID: PMC3763228 DOI: 10.1089/ars.2012.4736] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
AIMS Our previous work identified thioredoxin-like protein 2 (Txl-2) as the target of the monoclonal antibody MC3 associated with colon cancer, but its underlying mechanisms remain poorly understood. Txl-2, a novel thioredoxin (Trx) and nucleoside diphosphate kinase family member, is alternatively spliced and gives rise to three different Txl-2 isoforms. In this study, Txl-2 expression in colon cancer, differential functions for Txl-2 isoforms in cell invasion and metastasis, and the downstream signaling were investigated. RESULTS Txl-2 expression was elevated in colon cancer tissues compared to normal colonic tissues, with a high correlation between the histological grade and prognosis. Knockdown of Txl-2 expression significantly inhibited cancer cell motility, and the invasive and metastatic abilities of colon cancer cells. Interestingly, Txl-2 isoforms showed differential effects on cancer cell invasion and metastasis. Cell invasion and metastasis were significantly promoted by Txl-2b but inhibited by Txl-2c, while no obvious effect was observed for Txl-2a. Furthermore, a direct interaction was identified between Txl-2b and Ran, a Ras-related protein, by yeast two-hybrid assay and coimmunoprecipitation. PI3K pathway was found to be a major pathway mediating Txl-2b induced tumor invasion and metastasis. INNOVATION The current study provides a novel biomarker and target molecule for the diagnosis and treatment of colon cancer and provides a novel paradigm to understand how alternative splicing functions in human cancer. CONCLUSION Our findings demonstrate an elevated Txl-2 expression in colon cancer and that Txl-2b promotes cell invasion and metastasis through interaction with Ran and PI3K signaling pathway.
Collapse
Affiliation(s)
- Yuanyuan Lu
- State Key Laboratory of Cancer Biology, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
German P, Szaniszlo P, Hajas G, Radak Z, Bacsi A, Hazra TK, Hegde ML, Ba X, Boldogh I. Activation of cellular signaling by 8-oxoguanine DNA glycosylase-1-initiated DNA base excision repair. DNA Repair (Amst) 2013; 12:856-63. [PMID: 23890570 DOI: 10.1016/j.dnarep.2013.06.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 06/29/2013] [Accepted: 06/30/2013] [Indexed: 01/16/2023]
Abstract
Accumulation of 8-oxo-7,8-dihydroguanine (8-oxoG) in the DNA results in genetic instability and mutagenesis, and is believed to contribute to carcinogenesis, aging processes and various aging-related diseases. 8-OxoG is removed from the DNA via DNA base excision repair (BER), initiated by 8-oxoguanine DNA glycosylase-1 (OGG1). Our recent studies have shown that OGG1 binds its repair product 8-oxoG base with high affinity at a site independent from its DNA lesion-recognizing catalytic site and the OGG1•8-oxoG complex physically interacts with canonical Ras family members. Furthermore, exogenously added 8-oxoG base enters the cells and activates Ras GTPases; however, a link has not yet been established between cell signaling and DNA BER, which is the endogenous source of the 8-oxoG base. In this study, we utilized KG-1 cells expressing a temperature-sensitive mutant OGG1, siRNA ablation of gene expression, and a variety of molecular biological assays to define a link between OGG1-BER and cellular signaling. The results show that due to activation of OGG1-BER, 8-oxoG base is released from the genome in sufficient quantities for activation of Ras GTPase and resulting in phosphorylation of the downstream Ras targets Raf1, MEK1,2 and ERK1,2. These results demonstrate a previously unrecognized mechanism for cellular responses to OGG1-initiated DNA BER.
Collapse
Affiliation(s)
- Peter German
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
van den Berg MCW, van Gogh IJA, Smits AMM, van Triest M, Dansen TB, Visscher M, Polderman PE, Vliem MJ, Rehmann H, Burgering BMT. The small GTPase RALA controls c-Jun N-terminal kinase-mediated FOXO activation by regulation of a JIP1 scaffold complex. J Biol Chem 2013; 288:21729-41. [PMID: 23770673 DOI: 10.1074/jbc.m113.463885] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
FOXO (forkhead box O) transcription factors are tumor suppressors and increase the life spans of model organisms. Cellular stress, in particular oxidative stress caused by an increase in levels of reactive oxygen species (ROS), activates FOXOs through JNK-mediated phosphorylation. Importantly, JNK regulation of FOXO is evolutionarily conserved. Here we identified the pathway that mediates ROS-induced JNK-dependent FOXO regulation. Following increased ROS, RALA is activated by the exchange factor RLF (RalGDS-like factor), which is in complex with JIP1 (C-Jun-amino-terminal-interacting protein 1) and JNK. Active RALA consequently regulates assembly and activation of MLK3, MKK4, and JNK onto the JIP1 scaffold. Furthermore, regulation of FOXO by RALA and JIP1 is conserved in C. elegans, where both ral-1 and jip-1 depletion impairs heat shock-induced nuclear translocation of the FOXO orthologue DAF16.
Collapse
Affiliation(s)
- Maaike C W van den Berg
- Molecular Cancer Research, University Medical Center Utrecht, Utrecht, 3584 CG, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Corcoran A, Cotter TG. Redox regulation of protein kinases. FEBS J 2013; 280:1944-65. [PMID: 23461806 DOI: 10.1111/febs.12224] [Citation(s) in RCA: 226] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 02/24/2013] [Accepted: 02/27/2013] [Indexed: 12/30/2022]
Abstract
Reactive oxygen species (ROS) have been long regarded as by-products of a cascade of reactions stemming from cellular oxygen metabolism, which, if they accumulate to toxic levels, can have detrimental effects on cellular biomolecules. However, more recently, the recognition of ROS as mediators of cellular communications has led to their classification as signalling mediators in their own right. The prototypic redox-regulated targets downstream of ROS are the protein tyrosine phosphatases, and the wealth of research that has focused on this area has come to shape our understanding of how redox-signalling contributes to and facilitates protein tyrosine phosphorylation signalling cascades. However, it is becoming increasingly apparent that there is more to this system than simply the negative regulation of protein tyrosine phosphatases. Identification of redox-sensitive kinases such as Src led to the slow emergence of a role for redox regulation of tyrosine kinases. A flow of evidence, which has increased exponentially in recent times as a result of the development of new methods for the detection of oxidative modifications, demonstrates that, by concurrent oxidative activation of tyrosine kinases, ROS fine tune the duration and amplification of the phosphorylation signal. A more thorough understanding of the complex regulatory mechanism of redox-modification will allow targeting of both the production of ROS and their downstream effectors for therapeutic purposes. The present review assesses the most relevant recent literature that demonstrates a role for kinase regulation by oxidation, highlights the most significant findings and proposes future directions for this crucial area of redox biology.
Collapse
Affiliation(s)
- Aoife Corcoran
- Tumour Biology Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Ireland
| | | |
Collapse
|
42
|
The antagonistic roles of PDGF and integrin αvβ3 in regulating ROS production at focal adhesions. Biomaterials 2013; 34:3807-15. [PMID: 23465490 DOI: 10.1016/j.biomaterials.2013.01.092] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 01/26/2013] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) have been shown to play crucial roles in regulating various cellular functions, e.g. focal adhesion (FA) dynamics and cell migration upon growth factor stimulation. However, it is not clear how ROS are regulated at subcellular FA sites to impact cell migration. We have developed a biosensor capable of monitoring ROS production at FA sites in live cells with high sensitivity and specificity, utilizing fluorescence resonance energy transfer (FRET). The results revealed that platelet derived growth factor (PDGF) can induce ROS production at FA sites, which is mediated by Rac1 activation. In contrast, integrins, specifically integrin αvβ3, inhibits this local ROS production. The RhoA activity can mediate this inhibitory role of integrins in regulating ROS production. Therefore, PDGF and integrin αvβ3 coordinate to have an antagonistic effect in the ROS production at FA sites to regulate cell adhesion and migration.
Collapse
|
43
|
Mitchell L, Hobbs GA, Aghajanian A, Campbell SL. Redox regulation of Ras and Rho GTPases: mechanism and function. Antioxid Redox Signal 2013; 18:250-8. [PMID: 22657737 PMCID: PMC3518547 DOI: 10.1089/ars.2012.4687] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
SIGNIFICANCE Oxidation and reduction events are critical to physiological and pathological processes and are highly regulated. Herein, we present evidence for the role of Ras and Rho GTPases in controlling these events and the unique underlying mechanisms. Evidence for redox regulation of Ras GTPases that contain a redox-sensitive cysteine (X) in the conserved NKXD motif is presented, and a growing consensus supports regulation by a thiyl radical-mediated oxidation mechanism. We also discuss the debate within the literature regarding whether 2e(-) oxidation mechanisms also regulate Ras GTPase activity. RECENT ADVANCES We examine the increasing in vitro and cell-based data supporting oxidant-mediated activation of Rho GTPases that contain a redox-sensitive cysteine at the end of the conserved phosphoryl-binding loop (p-loop) motif (GXXXXG[S/T]C). While this motif is distinct from Ras, these data suggest a similar 1e(-) oxidation-mediated activation mechanism. CRITICAL ISSUES We also review the data showing that the unique p-loop placement of the redox-sensitive cysteine in Rho GTPases supports activation by 2e(-) cysteine oxidation. Finally, we examine the role that Ras and Rho GTPases play in controlling key oxidant-regulating enzymes in the cell, and we speculate on a feedback mechanism. FUTURE DIRECTIONS Given that these GTPases and redox-regulating enzymes are involved in multiple physiological and pathological processes, we discuss future experiments that may clarify the interplay between them.
Collapse
Affiliation(s)
- Lauren Mitchell
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | |
Collapse
|
44
|
Go YM, Duong DM, Peng J, Jones DP. Protein Cysteines Map to Functional Networks According to Steady-state Level of Oxidation. ACTA ACUST UNITED AC 2013; 4:196-209. [PMID: 22605892 DOI: 10.4172/jpb.1000190] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The cysteine (Cys) proteome serves critical roles in protein structure, function and regulation, and includes key targets in oxidative mechanisms of disease. Thioredoxins maintain Cys residues in thiol forms, and previous research shows that the redox potential of thioredoxin in mitochondria and nuclei is more reduced than cytoplasm, suggesting that proteins in these compartments may have different steady-state oxidation. This study measured fractional oxidation of 641 peptidyl Cys residues from 333 proteins in HT29 cells by mass spectrometry. Average oxidation of cytoplasmic, nuclear and mitochondrial proteins was similar (15.8, 15.5, 14%, respectively). Pathway analysis showed that more reduced cytoplasmic Cys were in proteins associated with the cytoskeleton, more reduced nuclear Cys with Ran signaling and RNA post-transcriptional modifcation, and more reduced mitochondrial Cys with energy metabolism, cell growth and cell proliferation. More oxidized cytoplasmic Cys included associations with PI3/Akt, Myc-mediated apoptosis and 14-3-3-mediated signaling. Weaker associations of oxidized nuclear and mitochondrial Cys occurred with granzyme B signaling and intermediary metabolism, respectively. Thus, steady-state peptidyl Cys oxidation is associated with functional pathways rather than simply with organellar distribution. This suggests that oxidative mechanisms of disease could target functional pathways or networks rather than individual proteins or subcellular compartments.
Collapse
Affiliation(s)
- Young-Mi Go
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory Proteomics Service Center, Emory University, Atlanta, Georgia 30322 USA
| | | | | | | |
Collapse
|
45
|
Abstract
Since the Central dogma of Molecular Biology was proposed about 40 years ago; our understanding of the intricacies of gene regulation has undergone tectonic shifts almost every decade. It is now widely accepted that the complexity of an organism is not directed by the sheer number of genes it carries but how they are decoded by a myriad of regulatory modules. Over the years, it has emerged that the organizations chromatins and its remodeling; splicing and polyadenylation of pre-mRNAs, stability and localization of mRNAs and modulation of their expression by non-coding and miRNAs play pivotal roles in metazoan gene expression. Nevertheless, in spite of tremendous progress in our understanding of all these mechanisms of gene regulation, the way these events are coordinated leading towards a highly defined proteome of a given cell type remains enigmatic. In that context, the structures of many metazoan genes cannot fully explain their pattern of expression in different tissues, especially during embryonic development and progression of various diseases. Further, numerous studies done during the past quarter of a century suggested that the heritable states of transcriptional activation or repression of a gene can be influenced by the covalent modifications of constituent bases and associated histones; its chromosomal context and long-range interactions between various chromosomal elements (Holliday 1987; Turner 1998; Lyon 1993). However, molecular dissection of these phenomena is largely unknown and is an exciting topic of research under the sub-discipline epigenetics (Gasser et al. 1998).
Collapse
Affiliation(s)
- Shyamal K Goswami
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India,
| |
Collapse
|
46
|
Reactive oxygen species are induced by Kaposi's sarcoma-associated herpesvirus early during primary infection of endothelial cells to promote virus entry. J Virol 2012; 87:1733-49. [PMID: 23175375 DOI: 10.1128/jvi.02958-12] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The entry of Kaposi's sarcoma-associated herpesvirus (KSHV) into human dermal microvascular endothelial cells (HMVEC-d), natural in vivo target cells, via macropinocytosis is initiated through a multistep process involving the binding of KSHV envelope glycoproteins with cell surface α3β1, αVβ3, and αVβ5 integrin molecules and tyrosine kinase ephrin-A2 receptor, followed by the activation of preexisting integrin-associated signaling molecules such as focal adhesion kinase (FAK), Src, c-Cbl, phosphoinositide 3-kinase (PI-3K), and Rho-GTPases. Many viruses, including KSHV, utilize cellular reactive oxygen species (ROS) for viral genomic replication and survival within host cells; however, the role of ROS in early events of viral entry and the induction of signaling has not been elucidated. Here we show that KSHV induced ROS production very early during the infection of HMVEC-d cells and that ROS production was sustained over the observation period (24 h postinfection). ROS induction was dependent on the binding of KSHV to the target cells, since pretreatment of the virus with heparin abolished ROS induction. Pretreatment of HMVEC-d cells with the antioxidant N-acetylcysteine (NAC) significantly inhibited KSHV entry, and consequently gene expression, without affecting virus binding. In contrast, H(2)O(2) treatment increased the levels of KSHV entry and infection. In addition, NAC inhibited KSHV infection-induced translocation of αVβ3 integrin into lipid rafts, actin-dependent membrane perturbations, such as blebs, observed during macropinocytosis, and activation of the signal molecules ephrin-A2 receptor, FAK, Src, and Rac1. In contrast, H(2)O(2) treatment increased the activation of ephrin-A2, FAK, Src, and Rac1. These studies demonstrate that KSHV infection induces ROS very early during infection to amplify the signaling pathways necessary for its efficient entry into HMVEC-d cells via macropinocytosis.
Collapse
|
47
|
Abstract
NADPH oxidases of the NADPH oxidase (NOX) family are dedicated reactive oxygen species-generating enzymes that broadly and specifically regulate redox-sensitive signalling pathways that are involved in cancer development and progression. They act at specific cellular membranes and microdomains through the activation of oncogenes and the inactivation of tumour suppressor proteins. In this Review, we discuss primary targets and redox-linked signalling systems that are influenced by NOX-derived ROS, and the biological role of NOX oxidases in the aetiology of cancer.
Collapse
Affiliation(s)
- Karen Block
- South Texas Veterans Health Care System, Audie L. Murphy Memorial Hospital Division, Department of Medicine, San Antonio, Texas 78229-73900, USA.
| | | |
Collapse
|
48
|
Garratt M, Brooks RC. Oxidative stress and condition-dependent sexual signals: more than just seeing red. Proc Biol Sci 2012; 279:3121-30. [PMID: 22648155 DOI: 10.1098/rspb.2012.0568] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The links between fitness, health, sexual signals and mate choice are complex and subject to ongoing study. In 1999, von Schantz et al. made the valuable suggestion that oxidative stress may be an important missing piece of this complex puzzle. Their suggestion has been enthusiastically tested, with over 300 studies citing their paper, but most effort has concerned carotenoid-based (and to a lesser extent melanin-based) visual signals, predominantly in birds and fishes. Today, we know a great deal more about oxidative stress and related physiology, in both a pathological and regulatory sense, than we did in 1999. We revisit von Schantz et al.'s predictions and, more importantly, highlight novel mechanisms that could link oxidative stress with a range of energetically demanding signals, greatly increasing the scope from visual signalling systems that are usually discussed and nearly always tested. In particular, we argue that differences between individuals in their ability to regulate physiology related to oxidative stress may be an important factor influencing the production of sexual signals and the costs that are incurred from investment.
Collapse
Affiliation(s)
- Michael Garratt
- Evolution and Ecology Research Centre, and School of Biological, Earth and Environmental Sciences, The University of New South Wales, Sydney, New South Wales 2052, Australia.
| | | |
Collapse
|
49
|
Abstract
The involvement of reactive oxygen species (ROS) in morphine-induced analgesia and tolerance has been suggested, yet how and where ROS take part in these processes remains largely unknown. Here, we report a novel role for the superoxide-generating enzyme NOX1/NADPH oxidase in the regulation of analgesia and acute analgesic tolerance. In mice lacking Nox1 (Nox1(-/Y)), the magnitude of the analgesia induced by morphine was significantly augmented. More importantly, analgesic tolerance induced by repeated administration of morphine was significantly suppressed compared with that in the littermates, wild-type Nox1(+/Y). In a membrane fraction obtained from the dorsal spinal cord, no difference was observed in morphine-induced [(35)S]GTPγS-binding between the genotypes, whereas morphine-stimulated GTPase activity was significantly attenuated in Nox1(-/Y). At 2 h after morphine administration, a significant decline in [(35)S]GTPγS-binding was observed in Nox1(+/Y) but not in Nox1(-/Y). No difference in the maximal binding and affinity of [(3)H]DAMGO was observed between the genotypes, but the translocation of protein kinase C isoforms to the membrane fraction following morphine administration was almost completely abolished in Nox1(-/Y). Finally, the phosphorylation of RGS9-2 and formation of a complex by Gαi2/RGS9-2 with 14-3-3 found in morphine-treated Nox1(+/Y) were significantly suppressed in Nox1(-/Y). Together, these results suggest that NOX1/NADPH oxidase attenuates the pharmacological effects of opioids by regulating GTPase activity and the phosphorylation of RGS9-2 by protein kinase C. NOX1/NADPH oxidase may thus be a novel target for the development of adjuvant therapy to retain the beneficial effects of morphine.
Collapse
|
50
|
Molecular Crosstalk between Integrins and Cadherins: Do Reactive Oxygen Species Set the Talk? JOURNAL OF SIGNAL TRANSDUCTION 2011; 2012:807682. [PMID: 22203898 PMCID: PMC3238397 DOI: 10.1155/2012/807682] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 08/24/2011] [Indexed: 11/18/2022]
Abstract
The coordinate modulation of the cellular functions of cadherins and integrins plays an essential role in fundamental physiological and pathological processes, including morphogenesis, tissue differentiation and renewal, wound healing, immune surveillance, inflammatory response, tumor progression, and metastasis. However, the molecular mechanisms underlying the fine-tuned functional communication between cadherins and integrins are still elusive. This paper focuses on recent findings towards the involvement of reactive oxygen species (ROS) in the regulation of cell adhesion and signal transduction functions of integrins and cadherins, pointing to ROS as emerging strong candidates for modulating the molecular crosstalk between cell-matrix and cell-cell adhesion receptors.
Collapse
|