1
|
Michalak KP, Michalak AZ. Understanding chronic inflammation: couplings between cytokines, ROS, NO, Ca i 2+, HIF-1α, Nrf2 and autophagy. Front Immunol 2025; 16:1558263. [PMID: 40264757 PMCID: PMC12012389 DOI: 10.3389/fimmu.2025.1558263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Chronic inflammation is an important component of many diseases, including autoimmune diseases, intracellular infections, dysbiosis and degenerative diseases. An important element of this state is the mainly positive feedback between inflammatory cytokines, reactive oxygen species (ROS), nitric oxide (NO), increased intracellular calcium, hypoxia-inducible factor 1-alpha (HIF-1α) stabilisation and mitochondrial oxidative stress, which, under normal conditions, enhance the response against pathogens. Autophagy and the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant response are mainly negatively coupled with the above-mentioned elements to maintain the defence response at a level appropriate to the severity of the infection. The current review is the first attempt to build a multidimensional model of cellular self-regulation of chronic inflammation. It describes the feedbacks involved in the inflammatory response and explains the possible pathways by which inflammation becomes chronic. The multiplicity of positive feedbacks suggests that symptomatic treatment of chronic inflammation should focus on inhibiting multiple positive feedbacks to effectively suppress all dysregulated elements including inflammation, oxidative stress, calcium stress, mito-stress and other metabolic disturbances.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | |
Collapse
|
2
|
Wei X, Huang Y, Sun C. A review of effects of electromagnetic fields on ageing and ageing dependent bioeffects of electromagnetic fields. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 963:178491. [PMID: 39818160 DOI: 10.1016/j.scitotenv.2025.178491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
Thanks to the progress of science and technology, human life expectancy has dramatically increased in the past few decades, but accompanied by rapid ageing of population, resulting in increased burden on society. At the same time, the living environment, especially the electromagnetic environment, has also greatly changed due to science and technology advances. The effect of artificial electromagnetic fields (EMFs) emitted from power lines, mobile phones, wireless equipment, and other devices on ageing and ageing-related diseases are receiving increasing attention. However, the information on the relationship between EMFs and ageing and ageing related susceptibility to EMFs is fragmentary, a review is needed. Only few studies directly investigate the effect of EMFs on ageing, and we reviewed the impact of EMFs on lifespan and cellular senescence to pry whether EMFs have an effect on ageing, and reviewed the age-dependent bioeffects and health impacts of EMFs to see whether ageing would affect biological susceptibility to EMFs. The results indicated that EMFs may have an effect on longevity and cellular senescence, but the results were inconsistent which may depend on EMF types (frequency, intensity, wave shape, etc.), species, and cell lines. Ageing has an impact on the biological or health effects of EMFs; however, the results differ depending on the EMF type and the endpoint or health outcome. Age-dependent changes in free radical metabolism, ion homeostasis, gene expression, enzyme activity, and tissue biophysical properties may be the reason; however, the underlying mechanisms are not fully elucidated.
Collapse
Affiliation(s)
- Xiaoxia Wei
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Yun Huang
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Chuan Sun
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital, 310030, Hangzhou, China.
| |
Collapse
|
3
|
Rasouli M, Fattahi R, Nuoroozi G, Zarei-Behjani Z, Yaghoobi M, Hajmohammadi Z, Hosseinzadeh S. The role of oxygen tension in cell fate and regenerative medicine: implications of hypoxia/hyperoxia and free radicals. Cell Tissue Bank 2024; 25:195-215. [PMID: 37365484 DOI: 10.1007/s10561-023-10099-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 06/18/2023] [Indexed: 06/28/2023]
Abstract
Oxygen pressure plays an integral role in regulating various aspects of cellular biology. Cell metabolism, proliferation, morphology, senescence, metastasis, and angiogenesis are some instances that are affected by different tensions of oxygen. Hyperoxia or high oxygen concentration, enforces the production of reactive oxygen species (ROS) that disturbs physiological homeostasis, and consequently, in the absence of antioxidants, cells and tissues are directed to an undesired fate. On the other side, hypoxia or low oxygen concentration, impacts cell metabolism and fate strongly through inducing changes in the expression level of specific genes. Thus, understanding the precise mechanism and the extent of the implication of oxygen tension and ROS in biological events is crucial to maintaining the desired cell and tissue function for application in regenerative medicine strategies. Herein, a comprehensive literature review has been performed to find out the impacts of oxygen tensions on the various behaviors of cells or tissues.
Collapse
Affiliation(s)
- Mehdi Rasouli
- Student Research Committee, Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Fattahi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Ghader Nuoroozi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Zeinab Zarei-Behjani
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maliheh Yaghoobi
- Engineering Department, Faculty of Chemical Engineering, Zanjan University, Zanjan, Iran
| | - Zeinab Hajmohammadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran
| | - Simzar Hosseinzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, 1985717443, Iran.
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Arora N, Keshri AK, Kaur R, Rawat SS, Kumar R, Mishra A, Prasad A. Taenia solium excretory secretory proteins (ESPs) suppresses TLR4/AKT mediated ROS formation in human macrophages via hsa-miR-125. PLoS Negl Trop Dis 2023; 17:e0011858. [PMID: 38157380 PMCID: PMC10783723 DOI: 10.1371/journal.pntd.0011858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 01/11/2024] [Accepted: 12/12/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Helminth infections are a global health menace affecting 24% of the world population. They continue to increase global disease burden as their unclear pathology imposes serious challenges to patient management. Neurocysticercosis is classified as neglected tropical disease and is caused by larvae of helminthic cestode Taenia solium. The larvae infect humans and localize in central nervous system and cause NCC; a leading etiological agent of acquired epilepsy in the developing world. The parasite has an intricate antigenic make-up and causes active immune suppression in the residing host. It communicates with the host via its secretome which is complex mixture of proteins also called excretory secretory products (ESPs). Understanding the ESPs interaction with host can identify therapeutic intervention hot spots. In our research, we studied the effect of T. solium ESPs on human macrophages and investigated the post-translation switch involved in its immunopathogenesis. METHODOLOGY T. solium cysts were cultured in vitro to get ESPs and used for treating human macrophages. These macrophages were studied for cellular signaling and miR expression and quantification at transcript and protein level. CONCLUSION We found that T. solium cyst ESPs treatment to human macrophages leads to activation of Th2 immune response. A complex cytokine expression by macrophages was also observed with both Th1 and Th2 cytokines in milieu. But, at the same time ESPs modulated the macrophage function by altering the host miR expression as seen with altered ROS activity, apoptosis and phagocytosis. This leads to activated yet compromised functional macrophages, which provides a niche to support parasite survival. Thus T. solium secretome induces Th2 phenomenon in macrophages which may promote parasite's survival and delay their recognition by host immune system.
Collapse
Affiliation(s)
- Naina Arora
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Anand K. Keshri
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Rimanpreet Kaur
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Suraj S. Rawat
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| | - Rajiv Kumar
- Biotechnology Division, CSIR-Institute for Himalayan Bioresource Technology, Palampur, Himachal Pradesh, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Amit Prasad
- School of Biosciences and Bioengineering, Indian Institute of Technology Mandi, Mandi, Himachal Pradesh, India
| |
Collapse
|
5
|
Madsen RR, Toker A. PI3K signaling through a biochemical systems lens. J Biol Chem 2023; 299:105224. [PMID: 37673340 PMCID: PMC10570132 DOI: 10.1016/j.jbc.2023.105224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023] Open
Abstract
Following 3 decades of extensive research into PI3K signaling, it is now evidently clear that the underlying network does not equate to a simple ON/OFF switch. This is best illustrated by the multifaceted nature of the many diseases associated with aberrant PI3K signaling, including common cancers, metabolic disease, and rare developmental disorders. However, we are still far from a complete understanding of the fundamental control principles that govern the numerous phenotypic outputs that are elicited by activation of this well-characterized biochemical signaling network, downstream of an equally diverse set of extrinsic inputs. At its core, this is a question on the role of PI3K signaling in cellular information processing and decision making. Here, we review the determinants of accurate encoding and decoding of growth factor signals and discuss outstanding questions in the PI3K signal relay network. We emphasize the importance of quantitative biochemistry, in close integration with advances in single-cell time-resolved signaling measurements and mathematical modeling.
Collapse
Affiliation(s)
- Ralitsa R Madsen
- MRC-Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, Scotland, United Kingdom.
| | - Alex Toker
- Department of Pathology and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
6
|
Next-Generation Sequencing Advances the Genetic Diagnosis of Cerebral Cavernous Malformation (CCM). Antioxidants (Basel) 2022; 11:antiox11071294. [PMID: 35883785 PMCID: PMC9311989 DOI: 10.3390/antiox11071294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/24/2022] [Accepted: 06/26/2022] [Indexed: 02/07/2023] Open
Abstract
Cerebral Cavernous Malformation (CCM) is a cerebrovascular disease of genetic origin that predisposes to seizures, focal neurological deficits and fatal intracerebral hemorrhage. It may occur sporadically or in familial forms, segregating as an autosomal dominant condition with incomplete penetrance and highly variable expressivity. Its pathogenesis has been associated with loss-of-function mutations in three genes, namely KRIT1 (CCM1), CCM2 and PDCD10 (CCM3), which are implicated in defense mechanisms against oxidative stress and inflammation. Herein, we screened 21 Italian CCM cases using clinical exome sequencing and found six cases (~29%) with pathogenic variants in CCM genes, including a large 145−256 kb genomic deletion spanning the KRIT1 gene and flanking regions, and the KRIT1 c.1664C>T variant, which we demonstrated to activate a donor splice site in exon 16. The segregation of this cryptic splicing mutation was studied in a large Italian family (five affected and seven unaffected cases), and showed a largely heterogeneous clinical presentation, suggesting the implication of genetic modifiers. Moreover, by analyzing ad hoc gene panels, including a virtual panel of 23 cerebrovascular disease-related genes (Cerebro panel), we found two variants in NOTCH3 and PTEN genes, which could contribute to the abnormal oxidative stress and inflammatory responses to date implicated in CCM disease pathogenesis.
Collapse
|
7
|
Kitching M, Inguva S, Ramani M, Gao Y, Marsili E, Cahill P. Biosynthesis of Gold Nanoparticles by Vascular Cells in vitro. Front Microbiol 2022; 13:813511. [PMID: 35479633 PMCID: PMC9036376 DOI: 10.3389/fmicb.2022.813511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/14/2022] [Indexed: 12/28/2022] Open
Abstract
Biosynthesis of gold nanoparticles (AuNPs) for antimicrobial and chemotherapeutic applications is a well-established process in microbial hosts such as bacterial, fungi, and plants. However, reports on AuNPs biosynthesis in mammalian cells are scarce. In this study, bovine aortic endothelial cells (BAECs) and bovine aortic smooth muscle cells (BASMCs) were examined for their ability to synthesize AuNPs in vitro. Cell culture conditions such as buffer selection, serum concentration, and HAuCl4 concentration were optimized before the biosynthesized AuNPs were characterized through visible spectrometry, transmission electron microscopy, X-ray diffraction, and Fourier transform infrared (FTIR) spectroscopy. BAECs and BASMC produced small, spherical AuNPs that are semi-crystalline with a similar diameter (23 ± 2 nm and 23 ± 4 nm). Hydrogen peroxide pretreatment increased AuNPs synthesis, suggesting that antioxidant enzymes may reduce Au3+ ions as seen in microbial cells. However, buthionine sulfoximine inhibition of glutathione synthesis, a key regulator of oxidative stress, failed to affect AuNPs generation. Taken together, these results show that under the right synthesis conditions, non-tumor cell lines can produce detectable concentrations of AuNPs in vitro.
Collapse
Affiliation(s)
- Michael Kitching
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, United States
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | - Meghana Ramani
- Nanotechnology Innovation Center of Kansas State, Department of Radiation Oncology, Wayne State University, Detroit, MI, United States
| | - Yina Gao
- Materials and Surface Science Institute, University of Limerick, Limerick, Ireland
| | - Enrico Marsili
- Department of Chemical and Materials Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Paul Cahill
- Vascular Biology and Therapeutics Laboratory, School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
8
|
Sindona C, Schepici G, Contestabile V, Bramanti P, Mazzon E. NOX2 Activation in COVID-19: Possible Implications for Neurodegenerative Diseases. ACTA ACUST UNITED AC 2021; 57:medicina57060604. [PMID: 34208136 PMCID: PMC8230853 DOI: 10.3390/medicina57060604] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a rapidly spreading contagious infectious disease caused by the pathogen severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that primarily affects the respiratory tract as well as the central nervous system (CNS). SARS-CoV-2 infection occurs through the interaction of the viral protein Spike with the angiotensin II receptor (ACE 2), leading to an increase of angiotensin II and activation of nicotinamide adenine dinucleotide phosphate oxidase2 (NOX2), resulting in the release of both reactive oxygen species (ROS) and inflammatory molecules. The purpose of the review is to explain that SARS-CoV-2 infection can determine neuroinflammation that induces NOX2 activation in microglia. To better understand the role of NOX2 in inflammation, an overview of its involvement in neurodegenerative diseases (NDs) such as Parkinson’s disease (PD), Alzheimer’s disease (AD), and amyotrophic lateral sclerosis (ALS) is provided. To write this manuscript, we performed a PubMed search to evaluate the possible relationship of SARS-CoV-2 infection in NOX2 activation in microglia, as well as the role of NOX2 in NDs. Several studies highlighted that NOX2 activation in microglia amplifies neuroinflammation. To date, there is no clinical treatment capable of counteracting its activation, however, NOX2 could be a promising pharmaceutical target useful for both the treatment and prevention of NDs and COVID-19 treatment.
Collapse
|
9
|
Ammendola R, Parisi M, Esposito G, Cattaneo F. Pro-Resolving FPR2 Agonists Regulate NADPH Oxidase-Dependent Phosphorylation of HSP27, OSR1, and MARCKS and Activation of the Respective Upstream Kinases. Antioxidants (Basel) 2021; 10:antiox10010134. [PMID: 33477989 PMCID: PMC7835750 DOI: 10.3390/antiox10010134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Formyl peptide receptor 2 (FPR2) is involved in the pathogenesis of chronic inflammatory diseases, being activated either by pro-resolving or proinflammatory ligands. FPR2-associated signal transduction pathways result in phosphorylation of several proteins and in NADPH oxidase activation. We, herein, investigated molecular mechanisms underlying phosphorylation of heat shock protein 27 (HSP27), oxidative stress responsive kinase 1 (OSR1), and myristolated alanine-rich C-kinase substrate (MARCKS) elicited by the pro-resolving FPR2 agonists WKYMVm and annexin A1 (ANXA1). Methods: CaLu-6 cells or p22phoxCrispr/Cas9 double nickase CaLu-6 cells were incubated for 5 min with WKYMVm or ANXA1, in the presence or absence of NADPH oxidase inhibitors. Phosphorylation at specific serine residues of HSP27, OSR1, and MARCKS, as well as the respective upstream kinases activated by FPR2 stimulation was analysed. Results: Blockade of NADPH oxidase functions prevents WKYMVm- and ANXA1-induced HSP-27(Ser82), OSR1(Ser339) and MARCKS(Ser170) phosphorylation. Moreover, NADPH oxidase inhibitors prevent WKYMVm- and ANXA1-dependent activation of p38MAPK, PI3K and PKCδ, the kinases upstream to HSP-27, OSR1 and MARCKS, respectively. The same results were obtained in p22phoxCrispr/Cas9 cells. Conclusions: FPR2 shows an immunomodulatory role by regulating proinflammatory and anti-inflammatory activities and NADPH oxidase is a key regulator of inflammatory pathways. The activation of NADPH oxidase-dependent pro-resolving downstream signals suggests that FPR2 signalling and NADPH oxidase could represent novel targets for inflammation therapeutic intervention.
Collapse
Affiliation(s)
| | | | | | - Fabio Cattaneo
- Correspondence: ; Tel.: +39-081-746-2036; Fax: +39-081-746-4359
| |
Collapse
|
10
|
Tauffenberger A, Magistretti PJ. Reactive Oxygen Species: Beyond Their Reactive Behavior. Neurochem Res 2021; 46:77-87. [PMID: 33439432 PMCID: PMC7829243 DOI: 10.1007/s11064-020-03208-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/02/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022]
Abstract
Cellular homeostasis plays a critical role in how an organism will develop and age. Disruption of this fragile equilibrium is often associated with health degradation and ultimately, death. Reactive oxygen species (ROS) have been closely associated with health decline and neurological disorders, such as Alzheimer's disease or Parkinson's disease. ROS were first identified as by-products of the cellular activity, mainly mitochondrial respiration, and their high reactivity is linked to a disruption of macromolecules such as proteins, lipids and DNA. More recent research suggests more complex function of ROS, reaching far beyond the cellular dysfunction. ROS are active actors in most of the signaling cascades involved in cell development, proliferation and survival, constituting important second messengers. In the brain, their impact on neurons and astrocytes has been associated with synaptic plasticity and neuron survival. This review provides an overview of ROS function in cell signaling in the context of aging and degeneration in the brain and guarding the fragile balance between health and disease.
Collapse
Affiliation(s)
- Arnaud Tauffenberger
- King Abdullah University of Science and Technology, Thuwal, 23955, Kingdom of Saudi Arabia.
| | - Pierre J Magistretti
- King Abdullah University of Science and Technology, Thuwal, 23955, Kingdom of Saudi Arabia.
| |
Collapse
|
11
|
Al-Azzam N, Teegala LR, Pokhrel S, Ghebreigziabher S, Chachkovskyy T, Thodeti S, Gavilanes I, Covington K, Thodeti CK, Paruchuri S. Transient Receptor Potential Vanilloid channel regulates fibroblast differentiation and airway remodeling by modulating redox signals through NADPH Oxidase 4. Sci Rep 2020; 10:9827. [PMID: 32555397 PMCID: PMC7299963 DOI: 10.1038/s41598-020-66617-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/22/2020] [Indexed: 12/31/2022] Open
Abstract
Asthma is characterized by pathological airway remodeling resulting from persistent myofibroblast activation. Although transforming growth factor beta 1 (TGFβ1), mechanical signals, and reactive oxygen species (ROS) are implicated in fibroblast differentiation, their integration is still elusive. We identified that Transient Receptor Potential Vanilloid 4 (TRPV4), a mechanosensitive ion channel mediates lung fibroblast (LF) differentiation and D. farinae-induced airway remodeling via a novel TRPV4-NADPH Oxidase 4 (NOX4) interaction. NOX4-mediated ROS production is essential for TGFβ1-induced LF differentiation via myocardin-related transcription factor-A (MRTF-A) and plasminogen activator inhibitor 1 (PAI-1). Importantly, TRPV4 inhibition prevented TGFβ1-induced NOX4 expression and ROS production. Both TRPV4 and NOX4 are activated by phosphatidylinositol 3-kinase (PI3K) downstream of TGFβ1, and signals from both TRPV4 and Rac are necessary for NOX4 upregulation. Notably, NOX4 expression is higher in fibroblasts derived from asthmatic patients (disease human LF; DHLF) in comparison to non-asthmatics (normal human LF; NHLF). Further, NOX4 expression is up-regulated in the lungs of D.farinae-treated wild type mice (WT) relative to saline-treated WT, which was attenuated in TRPV4 knockout (KO) mice. Our findings suggest that TRPV4 integrates TGFβ1 and ROS signaling through NOX4 and, TRPV4-NOX4 interaction is amenable to target lung remodeling during asthma.
Collapse
Affiliation(s)
- Nosayba Al-Azzam
- Department of Chemistry, University of Akron, Akron, OH, US.,Department of Physiology and Biochemistry, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Sabita Pokhrel
- Department of Chemistry, University of Akron, Akron, OH, US
| | | | | | - Sathwika Thodeti
- Department of Chemistry, University of Akron, Akron, OH, US.,Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, US
| | | | | | - Charles K Thodeti
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, US
| | | |
Collapse
|
12
|
Graber K, Khan F, Glück B, Weigel C, Marzo S, Doshi H, Ehrhardt C, Heller R, Gräler M, Henke A. The role of sphingosine-1-phosphate signaling in HSV-1-infected human umbilical vein endothelial cells. Virus Res 2020; 276:197835. [DOI: 10.1016/j.virusres.2019.197835] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 01/14/2023]
|
13
|
The mTOR/GCLc/GSH Pathway Mediates the Dose-Dependent Bidirectional Regulation of ROS Induced by TiO 2NPs in Neurogenic Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019. [DOI: 10.1155/2019/7621561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Objective. The effect of TiO2NP exposure on the nervous system and the underlying mechanism remain unclear. The antioxidant effect of TiO2NPs at a low dose was newly found in our study, which was different from the effect at high dose. This study is aimed at exploring the mechanism underlying the antioxidant effects of TiO2NPs at low dose and the induction of ROS accumulation by TiO2NPs at high dose in neurogenic cell lines.Methods. We measured the changes in key molecules in the ROS regulation pathway by western blotting, flow cytometry, and commercial assay kits, and these key molecules were further evaluated to verify their interactions and roles using SH-SY5Y, U251, and SK-N-SH cell lines treated with TiO2NPs.Results. Our results showed that the weak antioxidant effect at low dose was caused by mTOR/GCLc-induced GSH overproduction and GSH-Px activity impairment. ROS accumulation at high dose was caused by a mTOR/GCLc-mediated decrease in GSH production, GSH-Px activity impairment, and dramatic ROS production. Furthermore, we found that the ROS species were mainly O2-⋅, and that SOD played a crucial role in reducing O2-⋅levels before the mTOR protein was activated.Conclusion. We revealed the mechanism underlying the bidirectional regulation of ROS induced by TiO2NPs at different doses in neurogenic cell lines. Our study emphasized the potential neurotoxic effects of NPs at low dose, which should arouse concern about their safety.
Collapse
|
14
|
Knock GA. NADPH oxidase in the vasculature: Expression, regulation and signalling pathways; role in normal cardiovascular physiology and its dysregulation in hypertension. Free Radic Biol Med 2019; 145:385-427. [PMID: 31585207 DOI: 10.1016/j.freeradbiomed.2019.09.029] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
The last 20-25 years have seen an explosion of interest in the role of NADPH oxidase (NOX) in cardiovascular function and disease. In vascular smooth muscle and endothelium, NOX generates reactive oxygen species (ROS) that act as second messengers, contributing to the control of normal vascular function. NOX activity is altered in response to a variety of stimuli, including G-protein coupled receptor agonists, growth-factors, perfusion pressure, flow and hypoxia. NOX-derived ROS are involved in smooth muscle constriction, endothelium-dependent relaxation and smooth muscle growth, proliferation and migration, thus contributing to the fine-tuning of blood flow, arterial wall thickness and vascular resistance. Through reversible oxidative modification of target proteins, ROS regulate the activity of protein tyrosine phosphatases, kinases, G proteins, ion channels, cytoskeletal proteins and transcription factors. There is now considerable, but somewhat contradictory evidence that NOX contributes to the pathogenesis of hypertension through oxidative stress. Specific NOX isoforms have been implicated in endothelial dysfunction, hyper-contractility and vascular remodelling in various animal models of hypertension, pulmonary hypertension and pulmonary arterial hypertension, but also have potential protective effects, particularly NOX4. This review explores the multiplicity of NOX function in the healthy vasculature and the evidence for and against targeting NOX for antihypertensive therapy.
Collapse
Affiliation(s)
- Greg A Knock
- Dpt. of Inflammation Biology, School of Immunology & Microbial Sciences, Faculty of Life Sciences & Medicine, King's College London, UK.
| |
Collapse
|
15
|
Wu JR, You RI, Hu CT, Cheng CC, Rudy R, Wu WS. Hydrogen peroxide inducible clone-5 sustains NADPH oxidase-dependent reactive oxygen species-c-jun N-terminal kinase signaling in hepatocellular carcinoma. Oncogenesis 2019; 8:40. [PMID: 31387985 PMCID: PMC6684519 DOI: 10.1038/s41389-019-0149-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/18/2019] [Accepted: 06/21/2019] [Indexed: 12/19/2022] Open
Abstract
Target therapy aiming at critical molecules within the metastatic signal pathways is essential for prevention of hepatocellular carcinoma (HCC) progression. Hic-5 (hydrogen peroxide inducible clone-5) which belongs to the paxillin superfamily, can be stimulated by a lot of metastatic factors, such as transforming growth factor (TGF-β), hepatocyte growth factor (HGF), and reactive oxygen species (ROS). Previous studies implicated Hic-5 cross-talks with the ROS-c-jun N-terminal kinase (JNK) signal cascade in a positive feedback manner. In this report, we addressed this issue in a comprehensive manner. By RNA interference and ectopic Hic-5 expression, we demonstrated Hic-5 was essential for activation of NADPH oxidase and ROS generation leading to activation of downstream JNK and c-jun transcription factor. This was initiated by interaction of Hic-5 with the regulator and adaptor of NADPH oxidase, Rac1 and Traf4, respectively, which may further phosphorylate the nonreceptor tyrosine kinase Pyk2 at Tyr881. On the other hand, promoter activity assay coupled with deletion mapping and site directed mutagenesis strategies demonstrated the distal c-jun and AP4 putative binding regions (943–1126 bp upstream of translational start site) were required for transcriptional activation of Hic-5. Thus Hic-5 was both downstream and upstream of NADPH oxidase-ROS-JNK-c-jun cascade. This signal circuit was essential for regulating the expression of epithelial mesenchymal transition (EMT) factors, such as Snail, Zeb1, E-cadherin, and matrix metalloproteinase 9, involved in HCC cell migration and metastasis. Due to the limited expression of Hic-5 in normal tissue, it can be a promising therapeutic target for preventing HCC metastasis.
Collapse
Affiliation(s)
- Jia-Ru Wu
- Department of Molecular Biology and Human Genetics, Hualien, Taiwan
| | - Ren-In You
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Chi-Tan Hu
- Division of Gastroenterology, Department of Medicine, Buddhist Tzu Chi General Hospital and Tzu Chi University, Hualien, Taiwan.,Research Centre for Hepatology, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Chuan-Chu Cheng
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Rudy Rudy
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Sheng Wu
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, Taiwan. .,Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
16
|
Burtenshaw D, Kitching M, Redmond EM, Megson IL, Cahill PA. Reactive Oxygen Species (ROS), Intimal Thickening, and Subclinical Atherosclerotic Disease. Front Cardiovasc Med 2019; 6:89. [PMID: 31428618 PMCID: PMC6688526 DOI: 10.3389/fcvm.2019.00089] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/14/2019] [Indexed: 12/14/2022] Open
Abstract
Arteriosclerosis causes significant morbidity and mortality worldwide. Central to this process is the development of subclinical non-atherosclerotic intimal lesions before the appearance of pathologic intimal thickening and advanced atherosclerotic plaques. Intimal thickening is associated with several risk factors, including oxidative stress due to reactive oxygen species (ROS), inflammatory cytokines and lipid. The main ROS producing systems in-vivo are reduced nicotinamide dinucleotide phosphate (NADPH) oxidase (NOX). ROS effects are context specific. Exogenous ROS induces apoptosis and senescence, whereas intracellular ROS promotes stem cell differentiation, proliferation, and migration. Lineage tracing studies using murine models of subclinical atherosclerosis have revealed the contributory role of medial smooth muscle cells (SMCs), resident vascular stem cells, circulating bone-marrow progenitors and endothelial cells that undergo endothelial-mesenchymal-transition (EndMT). This review will address the putative physiological and patho-physiological roles of ROS in controlling vascular cell fate and ROS contribution to vascular regeneration and disease progression.
Collapse
Affiliation(s)
- Denise Burtenshaw
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| | | | - Eileen M Redmond
- Department of Surgery, University of Rochester, Rochester, NY, United States
| | - Ian L Megson
- Centre for Health Science, UHI Institute of Health Research and Innovation, Inverness, United Kingdom
| | - Paul A Cahill
- Vascular Biology & Therapeutics, School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
17
|
Lopes Pires ME, Antunes Naime AC, Oliveira JGF, Anhe GF, Garraud O, Cognasse F, Antunes E, Marcondes S. Signalling pathways involved in p47 phox -dependent reactive oxygen species in platelets of endotoxemic rats. Basic Clin Pharmacol Toxicol 2018; 124:394-403. [PMID: 30318767 DOI: 10.1111/bcpt.13148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 10/09/2018] [Indexed: 12/12/2022]
Abstract
Thrombocytopenia during sepsis is associated with a less favourable clinical outcome. Overproduction of reactive oxygen species (ROS) by different cell types contributes to sepsis. Platelets generate ROS, but the upstream pathways of NADPH oxidase activation are not completely understood. Here, we designed experiments in washed platelets from lipopolysaccharide (LPS)-treated rats to investigate the p47phox activation and ROS generation, and its modulation by c-Src family kinase (c-Src), phosphoinositide 3-kinase (PI3K), protein kinase C (PKC) and protein kinase G (PKG). Rats were injected intraperitoneally with LPS (1 mg/kg), and at 48 hours thereafter, arterial blood was collected and washed platelets were obtained. Washed platelets were pre-incubated with different inhibitors and subsequently activated or not with ADP. Flow cytometry, Western blotting and ELISA were performed. We found that LPS significantly increased the p47phox phosphorylation and ROS generation compared with the control group (P < 0.05). The enhanced ROS production in the LPS group was unaffected by the non-selective SFKs inhibitor PP2, the PI3K inhibitor wortmannin or the Akt inhibitor PPI-1. The cyclic GMP levels were 115% higher in activated platelets of LPS compared with the saline group (P < 0.05). Moreover, in the LPS group, the sGC inhibitor ODQ, the PKG inhibitor Rp-8-Br and the PKC inhibitor GF109203X abrogated the increased p47phox phosphorylation and reduced the ROS levels. In conclusion, selective inhibitors of cGMP-PKG and PKC-p47phox pathways that regulate ROS generation by LPS in platelets may help control the redox balance in sepsis improving the survival of patients.
Collapse
Affiliation(s)
- Maria E Lopes Pires
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ana C Antunes Naime
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Jessica G F Oliveira
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Gabriel F Anhe
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Oliver Garraud
- GIMAP-EA3064, Université de Lyon, Saint Etienne, France.,Etablissement Français du Sang (EFS) Rhône-Alpes-Auvergne, Saint-Etienne, France
| | - Fabrice Cognasse
- GIMAP-EA3064, Université de Lyon, Saint Etienne, France.,Institut National de Transfusion Sanguine (INTS), Paris, France
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Sisi Marcondes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
18
|
Koundouros N, Poulogiannis G. Phosphoinositide 3-Kinase/Akt Signaling and Redox Metabolism in Cancer. Front Oncol 2018; 8:160. [PMID: 29868481 PMCID: PMC5968394 DOI: 10.3389/fonc.2018.00160] [Citation(s) in RCA: 279] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 04/26/2018] [Indexed: 12/21/2022] Open
Abstract
Metabolic rewiring and the consequent production of reactive oxygen species (ROS) are necessary to promote tumorigenesis. At the nexus of these cellular processes is the aberrant regulation of oncogenic signaling cascades such as the phosphoinositide 3-kinase and AKT (PI3K/Akt) pathway, which is one of the most frequently dysregulated pathways in cancer. In this review, we examine the regulation of ROS metabolism in the context of PI3K-driven tumors with particular emphasis on four main areas of research. (1) Stimulation of ROS production through direct modulation of mitochondrial bioenergetics, activation of NADPH oxidases (NOXs), and metabolic byproducts associated with hyperactive PI3K/Akt signaling. (2) The induction of pro-tumorigenic signaling cascades by ROS as a consequence of phosphatase and tensin homolog and receptor tyrosine phosphatase redox-dependent inactivation. (3) The mechanisms through which PI3K/Akt activation confers a selective advantage to cancer cells by maintaining redox homeostasis. (4) Opportunities for therapeutically exploiting redox metabolism in PIK3CA mutant tumors and the potential for implementing novel combinatorial therapies to suppress tumor growth and overcome drug resistance. Further research focusing on the multi-faceted interactions between PI3K/Akt signaling and ROS metabolism will undoubtedly contribute to novel insights into the extensive pro-oncogenic effects of this pathway, and the identification of exploitable vulnerabilities for the treatment of hyperactive PI3K/Akt tumors.
Collapse
Affiliation(s)
- Nikos Koundouros
- Department of Cancer Biology, Institute of Cancer Research, London, United Kingdom.,Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - George Poulogiannis
- Department of Cancer Biology, Institute of Cancer Research, London, United Kingdom.,Division of Computational and Systems Medicine, Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
19
|
Nox, Reactive Oxygen Species and Regulation of Vascular Cell Fate. Antioxidants (Basel) 2017; 6:antiox6040090. [PMID: 29135921 PMCID: PMC5745500 DOI: 10.3390/antiox6040090] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 10/21/2017] [Accepted: 11/07/2017] [Indexed: 01/09/2023] Open
Abstract
The generation of reactive oxygen species (ROS) and an imbalance of antioxidant defence mechanisms can result in oxidative stress. Several pro-atherogenic stimuli that promote intimal-medial thickening (IMT) and early arteriosclerotic disease progression share oxidative stress as a common regulatory pathway dictating vascular cell fate. The major source of ROS generated within the vascular system is the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family of enzymes (Nox), of which seven members have been characterized. The Nox family are critical determinants of the redox state within the vessel wall that dictate, in part the pathophysiology of several vascular phenotypes. This review highlights the putative role of ROS in controlling vascular fate by promoting endothelial dysfunction, altering vascular smooth muscle phenotype and dictating resident vascular stem cell fate, all of which contribute to intimal medial thickening and vascular disease progression.
Collapse
|
20
|
Zheng Y, Li X, Manor LC, Cao H, Chen Q. An Integrative Computational Approach to Evaluate Genetic Markers for Chronic Lymphocytic Leukemia. J Comput Biol 2017; 24:942-952. [PMID: 28570130 PMCID: PMC11753456 DOI: 10.1089/cmb.2017.0041] [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] [Indexed: 11/12/2022] Open
Abstract
Recent studies reported hundreds of genes linked to chronic lymphocytic leukemia (CLL). However, many of these candidate genes were lack of replication and results were not always consistent. Here, we proposed a computational workflow to curate and evaluate CLL-related genes. The method integrates large-scale literature knowledge data, gene expression data, and related pathways/network information for quantitative marker evaluation. Pathway Enrichment, Sub-Network Enrichment, and Gene-Gene Interaction analysis were conducted to study the pathogenic profile of the candidate genes, with four metrics proposed and validated for each gene. By using our approach, a scalable CLL genetic database was developed including CLL-related genes, pathways, diseases and information of supporting references. The CLL case/control classification supported the effectiveness of the four proposed metrics, which successfully identified nine well-studied CLL genes (i.e., TNF, BCL2, TP53, VEGFA, P2RX7, AKT1, SYK, IL4, and MDM2) and highlighted two newly reported CLL genes (i.e., PDGFRA and CSF1R). The computational biology approach and the CLL database developed in this study provide a valuable resource that may facilitate the understanding of the genetic profile of CLL.
Collapse
Affiliation(s)
- Yu Zheng
- Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Xiaoyang Li
- Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Lydia C. Manor
- Department of Bioinformatics Service, American Informatics Consultant LLC, Rockville, Maryland
| | - Hongbao Cao
- Department of Genomics Research, R&D Solutions, Elsevier Inc., Rockville, Maryland
- Unit on Statistical Genomics, NIMH/NIH, Bethesda, Maryland
| | - Qiusheng Chen
- Department of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| |
Collapse
|
21
|
McGowan SE, McCoy DM. Platelet-derived growth factor receptor-α and Ras-related C3 botulinum toxin substrate-1 regulate mechano-responsiveness of lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2017; 313:L1174-L1187. [PMID: 28775097 DOI: 10.1152/ajplung.00185.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 12/23/2022] Open
Abstract
Platelet-derived growth factor (PDGF)-A, which only signals through PDGF-receptor-α (PDGFR-α), is required for secondary alveolar septal formation. Although PDGFR-α distinguishes mesenchymal progenitor cells during the saccular stage, PDGFR-α-expressing alveolar cells persist through adulthood. PDGF-A sustains proliferation, limits apoptosis, and maintains α-smooth muscle actin (α-SMA)-containing alveolar cells, which congregate at the alveolar entry ring at postnatal day (P)12. PDGFR-α-expressing, α-SMA-containing alveolar cells redistribute in the elongating septum, suggesting that they migrate to the alveolar entry rings, where mechanical tension is higher. We hypothesized that PDGFR-α and Ras-related C3 botulinum toxin substrate 1(Rac1) are required for mechanosensitive myofibroblast migration. Spreading of PDGFR-α-deficient lung fibroblasts was insensitive to increased rigidity, and their migration was not reduced by Rac1-guanine exchange factor (GEF)-inhibition. PDGFR-α-expressing fibroblasts migrated toward stiffer regions within two-dimensional substrates by increasing migrational persistence (durotaxis). Using a Förster resonance energy transfer (FRET) biosensor for Rac1-GTP, we observed that PDGFR-α was required for fibroblast Rac1 responsiveness to stiffness within a three-dimensional collagen substrate, which by itself increased Rac1-FRET. Rho-GTPase stabilized, whereas Rac1-GTPase increased the turnover of focal adhesions. Under conditions that increased Rac1-GTP, PDGFR-α signaled through both phosphoinositide-3-kinase (PIK) or Src to engage the Rac1 GEF dedicator of cytokinesis-1 (Dock180) and p21-activated-kinase interacting exchange factor-β (βPIX). In cooperation with collagen fibers, these signaling pathways may guide fibroblasts toward the more rigid alveolar entry ring during secondary septation. Because emphysema and interstitial fibrosis disrupt the parenchymal mechanical continuum, understanding how mechanical factors regulate fibroblast migration could elicit strategies for alveolar repair and regeneration.
Collapse
Affiliation(s)
- Stephen E McGowan
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Diann M McCoy
- Department of Veterans Affairs Research Service and Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
22
|
Viswambharan H, Yuldasheva NY, Sengupta A, Imrie H, Gage MC, Haywood N, Walker AM, Skromna A, Makova N, Galloway S, Shah P, Sukumar P, Porter KE, Grant PJ, Shah AM, Santos CX, Li J, Beech DJ, Wheatcroft SB, Cubbon RM, Kearney MT. Selective Enhancement of Insulin Sensitivity in the Endothelium In Vivo Reveals a Novel Proatherosclerotic Signaling Loop. Circ Res 2017; 120:784-798. [DOI: 10.1161/circresaha.116.309678] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 12/21/2022]
Abstract
Rationale:
In the endothelium, insulin stimulates endothelial NO synthase (eNOS) to generate the antiatherosclerotic signaling radical NO. Insulin-resistant type 2 diabetes mellitus is associated with reduced NO availability and accelerated atherosclerosis. The effect of enhancing endothelial insulin sensitivity on NO availability is unclear.
Objective:
To answer this question, we generated a mouse with endothelial cell (EC)–specific overexpression of the human insulin receptor (hIRECO) using the Tie2 promoter–enhancer.
Methods and Results:
hIRECO demonstrated significant endothelial dysfunction measured by blunted endothelium-dependent vasorelaxation to acetylcholine, which was normalized by a specific Nox2 NADPH oxidase inhibitor. Insulin-stimulated phosphorylation of protein kinase B was increased in hIRECO EC as was Nox2 NADPH oxidase–dependent generation of superoxide, whereas insulin-stimulated and shear stress–stimulated eNOS activations were blunted. Phosphorylation at the inhibitory residue Y657 of eNOS and expression of proline-rich tyrosine kinase 2 that phosphorylates this residue were significantly higher in hIRECO EC. Inhibition of proline-rich tyrosine kinase 2 improved insulin-induced and shear stress–induced eNOS activation in hIRECO EC.
Conclusions:
Enhancing insulin sensitivity specifically in EC leads to a paradoxical decline in endothelial function, mediated by increased tyrosine phosphorylation of eNOS and excess Nox2-derived superoxide. Increased EC insulin sensitivity leads to a proatherosclerotic imbalance between NO and superoxide. Inhibition of proline-rich tyrosine kinase 2 restores insulin-induced and shear stress–induced NO production. This study demonstrates for the first time that increased endothelial insulin sensitivity leads to a proatherosclerotic imbalance between NO and superoxide.
Collapse
Affiliation(s)
- Hema Viswambharan
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Nadira Y. Yuldasheva
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Anshuman Sengupta
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Helen Imrie
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Matthew C. Gage
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Natalie Haywood
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Andrew M.N. Walker
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Anna Skromna
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Natallia Makova
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Stacey Galloway
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Pooja Shah
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Piruthivi Sukumar
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Karen E. Porter
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Peter J. Grant
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Ajay M. Shah
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Celio X.C. Santos
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Jing Li
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - David J. Beech
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Stephen B. Wheatcroft
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Richard M. Cubbon
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| | - Mark T. Kearney
- From the Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, United Kingdom (H.V., N.Y.Y., A.S., H.I., N.H., A.M.N.W., A.S., N.M., S.G., P. Shah, P. Sukumar, K.E.P., P.J.G., J.L., D.J.B., S.B.W., R.M.C., M.T.K.); Division of Medicine, Department of Metabolism & Experimental Therapeutics, University College London, United Kingdom (M.C.G.); and British Heart Foundation Centre of Research Excellence, King’s College London, United Kingdom (A.M.S., C.X.C.S.)
| |
Collapse
|
23
|
Ferreira LF, Laitano O. Regulation of NADPH oxidases in skeletal muscle. Free Radic Biol Med 2016; 98:18-28. [PMID: 27184955 PMCID: PMC4975970 DOI: 10.1016/j.freeradbiomed.2016.05.011] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/31/2016] [Accepted: 05/12/2016] [Indexed: 12/20/2022]
Abstract
The only known function of NAD(P)H oxidases is to produce reactive oxygen species (ROS). Skeletal muscles express three isoforms of NAD(P)H oxidases (Nox1, Nox2, and Nox4) that have been identified as critical modulators of redox homeostasis. Nox2 acts as the main source of skeletal muscle ROS during contractions, participates in insulin signaling and glucose transport, and mediates the myocyte response to osmotic stress. Nox2 and Nox4 contribute to skeletal muscle abnormalities elicited by angiotensin II, muscular dystrophy, heart failure, and high fat diet. Our review addresses the expression and regulation of NAD(P)H oxidases with emphasis on aspects that are relevant to skeletal muscle. We also summarize: i) the most widely used NAD(P)H oxidases activity assays and inhibitors, and ii) studies that have defined Nox enzymes as protagonists of skeletal muscle redox homeostasis in a variety of health and disease conditions.
Collapse
Affiliation(s)
- Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA.
| | - Orlando Laitano
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, USA; Universidade Federal do Vale do São Francisco, Petrolina, PE, Brazil
| |
Collapse
|
24
|
Coughlan MT, Sharma K. Challenging the dogma of mitochondrial reactive oxygen species overproduction in diabetic kidney disease. Kidney Int 2016; 90:272-279. [PMID: 27217197 DOI: 10.1016/j.kint.2016.02.043] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/21/2016] [Accepted: 02/24/2016] [Indexed: 01/02/2023]
Abstract
The paradigm that high glucose drives overproduction of superoxide from mitochondria as a unifying theory to explain end organ damage in diabetes complications has been tightly held for more than a decade. With the recent development of techniques and probes to measure the production of distinct reactive oxygen species (ROS) in vivo, this widely held dogma is now being challenged with the emerging view that specific ROS moieties are essential for the function of specific intracellular signaling pathways and represent normal mitochondrial function. This review will provide a balanced overview of the dual nature of ROS, detailing current evidence for ROS overproduction in diabetic kidney disease, with a focus on cell types and sources of ROS. The technical aspects of measurement of mitochondrial ROS, both in isolated mitochondria and emerging in vivo methods will be discussed. The counterargument, that mitochondrial ROS production is reduced in diabetic complications, is consistent with a growing recognition that stimulation of mitochondrial biogenesis and oxidative phosphorylation activity reduces inflammation and fibrosis. It is clear that there is an urgent need to fully characterize ROS production paying particular attention to spatiotemporal aspects and to factor in the relevance of ROS in the regulation of cellular signaling in the pathogenesis of diabetic kidney disease. With improved tools and real-time imaging capacity, a greater understanding of the complex role of ROS will be able to guide novel therapeutic regimens.
Collapse
Affiliation(s)
- Melinda T Coughlan
- Baker International Diabetes Institute (IDI) Heart and Diabetes Institute, Melbourne, Victoria, Australia; Department of Medicine, Central Clinical School, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia; Department of Epidemiology and Preventive Medicine, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia
| | - Kumar Sharma
- Center for Renal Translational Medicine, Division of Nephrology-Hypertension, Institute of Metabolomic Medicine, University of California-San Diego, La Jolla, California, USA; Division of Medical Genetics, Department of Medicine, University of California-San Diego, La Jolla, California, USA; Division of Nephrology-Hypertension, Veterans Affairs San Diego Healthcare System, La Jolla, California, USA.
| |
Collapse
|
25
|
Wu YM, Chen ZJ, Jiang GM, Zhang KS, Liu Q, Liang SW, Zhou Y, Huang HB, Du J, Wang HS. Inverse agonist of estrogen-related receptor α suppresses the growth of triple negative breast cancer cells through ROS generation and interaction with multiple cell signaling pathways. Oncotarget 2016; 7:12568-81. [PMID: 26871469 PMCID: PMC4914305 DOI: 10.18632/oncotarget.7276] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/23/2016] [Indexed: 12/19/2022] Open
Abstract
There is an urgent clinical need for targeted therapy approaches for triple-negative breast cancer (TNBC) patients. Increasing evidences suggested that the expression of estrogen-related receptor alpha (ERRα) was correlate with unfavorable clinical outcomes of breast cancer patients. We here show that inhibition of ERRα by its inverse agonist XCT-790 can suppress the proliferation, decrease G2/M phases, and induce mitochondrial-related apoptosis of TNBC cells. XCT-790 elevates the proteins related to endoplasmic reticulum (ER) stress such as ATF4/6, XBT-1 and CHOP. It also increases the expression of growth inhibition related proteins such as p53 and p21. Further, XCT-790 can increase the generation of reactive oxygen species (ROS) in TNBC cells mainly through inhibition of SOD1/2. While ROS scavenger NAC abolishes XCT-790 induced ER-stress and growth arrest. XCT-790 treatment can rapidly activate the signal molecules including ERK1/2, p38-MAPK, JNK, Akt, p65, and IκBα, while NAC attenuates effects of XCT-790 induced phosphorylation of ERK1/2, p38-MAPK and Akt. Further, the inhibitors of ERK1/2, JNK, Akt, and NF-κB attenuate XCT-790 induced ROS generation. These data suggest that AKT/ROS and ERK/ROS positive feedback loops, NF-κB/ROS, and ROS/p38-MAPK, are activated in XCT-790 treated TNBC cells. In vivo experiments show that XCT-790 significantly suppresses the growth of MDA-MB-231 xenograft tumors, which is associated with up regulation of p53, p21, ER-stress related proteins while down regulation of bcl-2. The present discovery makes XCT-790 a promising candidate drug and lays the foundation for future development of ERRα-based therapies for TNBC patients.
Collapse
Affiliation(s)
- Ying-Min Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhuo-Jia Chen
- Department of Pharmacy, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Guan-Min Jiang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Kun-Shui Zhang
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Qiao Liu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Shu-Wei Liang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yan Zhou
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hong-Bin Huang
- Department of Pharmacy, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
26
|
Suppression of Nkx3.2 by phosphatidylinositol-3-kinase signaling regulates cartilage development by modulating chondrocyte hypertrophy. Cell Signal 2015; 27:2389-400. [PMID: 26363466 DOI: 10.1016/j.cellsig.2015.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 09/07/2015] [Indexed: 01/04/2023]
Abstract
Phosphatidylinositol-3-kinase (PI3K) is a key regulator of diverse biological processes including cell proliferation, migration, survival, and differentiation. While a role of PI3K in chondrocyte differentiation has been suggested, its precise mechanisms of action are poorly understood. Here we show that PI3K signaling can down-regulate Nkx3.2 at both mRNA and protein levels in various chondrocyte cultures in vitro. In addition, we have intriguingly found that p85β, not p85α, is specifically employed as a regulatory subunit for PI3K-mediated Nkx3.2 suppression. Furthermore, we found that regulation of Nkx3.2 by PI3K requires Rac1-PAK1, but not Akt, signaling downstream of PI3K. Finally, using embryonic limb bud cultures, ex vivo long bone cultures, and p85β knockout mice, we demonstrated that PI3K-mediated suppression of Nkx3.2 in chondrocytes plays a role in the control of cartilage hypertrophy during skeletal development in vertebrates.
Collapse
|
27
|
Liu LZ, Ding M, Zheng JZ, Zhu Y, Fenderson BA, Li B, Yu JJ, Jiang BH. Tungsten Carbide-Cobalt Nanoparticles Induce Reactive Oxygen Species, AKT, ERK, AP-1, NF-κB, VEGF, and Angiogenesis. Biol Trace Elem Res 2015; 166:57-65. [PMID: 25893364 DOI: 10.1007/s12011-015-0331-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 04/06/2015] [Indexed: 01/28/2023]
Abstract
Powder mixtures of tungsten carbide and metallic cobalt (WC-Co) are widely used in various products. Nanoparticles are engineered structures with at least one dimension of 100 nm or smaller. WC-Co is known to be associated with lung injury and diseases. Angiogenesis is a key process during vasculature, carcinogenesis, recovery of injury, and inflammatory diseases. However, the cellular effects of WC-Co nanoparticles on angiogenesis remain to be elucidated. In this study, we investigated angiogenic response and relative mechanisms after exposure to WC-Co nanoparticles. Our results showed that WC-Co nanoparticles at 5 μg/cm(2) induced ROS production which activated AKT and ERK1/2 signaling pathways in lung epithelial cells by reactive oxygen species (ROS) staining and immunoblotting; WC-Co treatment also increased transcriptional activation of AP-1, NF-κB, and VEGF by reporter assay. Further studies demonstrated that ROS are upstream molecules of AKT and ERK signaling pathways; the activation of AP-1, NF-κB, and VEGF was through ROS generation, AKT and ERK1/2 activation. In addition, WC-Co nanoparticles affected the cells to induce angiogenesis by chicken chorioallantoic membrane (CAM) assay. These results illustrate that exposure to WC-Co nanoparticles induces angiogenic response by activating ROS, AKT, and ERK1/2 signaling pathways and the downstream molecules and elucidate the potential molecular mechanisms during this process. This information may be useful for preventing potential damage from nanoparticle exposure in the future.
Collapse
Affiliation(s)
- Ling-Zhi Liu
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA,
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Ibrahim BM, Fan M, Abdel-Rahman AA. Oxidative stress and autonomic dysregulation contribute to the acute time-dependent myocardial depressant effect of ethanol in conscious female rats. Alcohol Clin Exp Res 2014; 38:1205-15. [PMID: 24754626 DOI: 10.1111/acer.12363] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 12/23/2013] [Indexed: 12/01/2022]
Abstract
BACKGROUND The molecular mechanisms of the acute hypotensive and indirectly assessed cardiac depressant effect of ethanol (EtOH)-evoked myocardial depression and hypotension in female rats are not known. We tested the hypothesis that a time-dependent myocardial depression caused by EtOH is initiated by its direct and indirect (cardiac vagal dominance) effects and is exacerbated by gradual development of oxidative stress. METHODS In conscious female rats, we directly measured left ventricular developed pressure (LVDP), the maximal rise of ventricular pressure over time (dP/dtmax ), blood pressure (BP), heart rate (HR), and sympathovagal activity following intragastric EtOH (1 g/kg) or water over 90 minutes. Catalytic activity of acetaldehyde (ACA)-generating (alcohol dehydrogenase [ADH] and catalase) and eliminating aldehyde dehydrogenase [ALDH2] enzymes along with mediators of oxidative stress were measured in myocardial tissues collected at 30, 60, or 90 minutes after EtOH or water. RESULTS EtOH reduced myocardial function (LVDP and dP/dtmax ) within 5 to 10 minutes before the steady fall in BP in conscious proestrus rats. Further, EtOH shifted the sympathovagal balance, analyzed by spectral analysis of high frequency and low frequency of interbeat intervals, toward vagal dominance. Prior vagal blockade (atropine) or antioxidant (tempol) treatment attenuated EtOH-evoked myocardial depression and hypotension. Ex vivo studies revealed time-dependent: (i) enhancement of ADH, but not ALDH2 activity (indicative of elevated ACA levels), (ii) increases in phosphorylated Akt and ERK1/2, NADPH-oxidase activity, reactive oxygen species, malondialdehyde, and 4-hydroxy-2-nonenal-modified proteins. These molecular responses along with reduced myocardial catalase activity were most evident at 90 minutes post-EtOH when the reductions in cardiac function and BP reached their nadir. CONCLUSIONS Vagal dominance and time-dependent myocardial oxidative stress along with the accumulation of cardiotoxic aldehydes mediate EtOH-evoked myocardial dysfunction and hypotension in conscious proestrus female rats.
Collapse
Affiliation(s)
- Badr M Ibrahim
- Department of Pharmacology and Toxicology (BMI, MF, AAR-R), Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | | | | |
Collapse
|
29
|
Ham SA, Lee H, Hwang JS, Kang ES, Yoo T, Paek KS, Do JT, Park C, Oh JW, Kim JH, Han CW, Seo HG. Activation of Peroxisome Proliferator-Activated Receptor δ Inhibits Angiotensin II-Induced Activation of Matrix Metalloproteinase-2 in Vascular Smooth Muscle Cells. J Vasc Res 2014; 51:221-30. [DOI: 10.1159/000365250] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 06/07/2014] [Indexed: 11/19/2022] Open
|
30
|
Chung KJ, Mitroulis I, Wiessner JR, Zheng YY, Siegert G, Sperandio M, Chavakis T. A novel pathway of rapid TLR-triggered activation of integrin-dependent leukocyte adhesion that requires Rap1 GTPase. Mol Biol Cell 2014; 25:2948-55. [PMID: 25057020 PMCID: PMC4230584 DOI: 10.1091/mbc.e14-04-0867] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
TLR2 and TLR5 ligation directly induces β2-integrin activation, promoting cell adhesion to ICAM-1. Systemic in vivo administration of the TLR2 ligand Pam3CSK4 increases integrin-dependent adhesion to endothelium within minutes. The signaling pathway linking TLR ligation with β2-integin activation involves Rac-1, NADPH oxidase 2, and Rap1-GTPase. Rapid β2-integrin activation is indispensable for leukocyte adhesion and recruitment to sites of infection and is mediated by chemokine- or P-selectin glycoprotein ligand-1–induced inside-out signaling. Here we uncovered a novel pathway for rapid activation of integrin-dependent leukocyte adhesion, triggered by toll-like receptor (TLR)–mediated signaling. TLR2 or TLR5 ligation rapidly activated integrin-dependent leukocyte adhesion to immobilized ICAM-1 and fibronectin. Consistently, in vivo administration of the TLR2-ligand Pam3CSK4 increased integrin-dependent slow rolling and adhesion to endothelium within minutes, as identified by intravital microscopy in the cremaster model. TLR2 and TLR5 ligation increased β2-integrin affinity, as assessed by the detection of activation-dependent neoepitopes. TLR2- and TLR5-triggered integrin activation in leukocytes required enhanced Rap1 GTPase activity, which was mediated by Rac1 activation and NADPH oxidase-2–dependent reactive oxygen species production. This novel direct pathway linking initial pathogen recognition by TLRs to rapid β2-integrin activation may critically regulate acute leukocyte infiltration to sites of pathogen invasion.
Collapse
Affiliation(s)
- Kyoung-Jin Chung
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, 01309 Dresden, Germany Institute of Physiology, Technische Universität Dresden, 01309 Dresden, Germany
| | - Ioannis Mitroulis
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, 01309 Dresden, Germany Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01309 Dresden, Germany
| | - Johannes R Wiessner
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians Universität, 80539 Munich, Germany
| | - Ying Yi Zheng
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL 32610
| | - Gabriele Siegert
- Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01309 Dresden, Germany
| | - Markus Sperandio
- Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians Universität, 80539 Munich, Germany
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry, Technische Universität Dresden, 01309 Dresden, Germany Institute of Physiology, Technische Universität Dresden, 01309 Dresden, Germany Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, 01309 Dresden, Germany Department of Medicine III, Technische Universität Dresden, 01309 Dresden, Germany
| |
Collapse
|
31
|
Htet Hlaing K, Clément MV. Formation of protein S-nitrosylation by reactive oxygen species. Free Radic Res 2014; 48:996-1010. [DOI: 10.3109/10715762.2014.942842] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
32
|
Nakanishi A, Wada Y, Kitagishi Y, Matsuda S. Link between PI3K/AKT/PTEN Pathway and NOX Proteinin Diseases. Aging Dis 2014; 5:203-11. [PMID: 24900943 DOI: 10.14336/ad.2014.0500203] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 10/27/2013] [Accepted: 10/30/2013] [Indexed: 12/17/2022] Open
Abstract
Accumulating evidence has revealed that thePI3K/AKT/PTENpathway acts as a pivotal determinant of cell fate regarding senescence and apoptosis, which is mediated by intracellular reactive oxygen species (ROS) generation. NADPH oxidase (NOX) family of enzymes generates the ROS. The regulation of NOX enzymes is complex, with many members of this family exhibiting complexity in terms of subunit composition, cellular location, and tissue-specific expression. Cells are continuously exposed to the ROS, which represent mutagens and are thought to be a major contributor to several diseases including cancer and aging process. Therefore, cellular ROS sensing and metabolism are firmly regulated by a variety of proteins involved in the redox mechanism. In this review, the roles of oxidative stress in PI3K/AKT/PTEN signaling are summarized with a focus on the links between the pathways and NOX protein in several diseases including cancer and aging.
Collapse
Affiliation(s)
| | - Yoko Wada
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
33
|
Frijhoff J, Dagnell M, Godfrey R, Ostman A. Regulation of protein tyrosine phosphatase oxidation in cell adhesion and migration. Antioxid Redox Signal 2014; 20:1994-2010. [PMID: 24111825 DOI: 10.1089/ars.2013.5643] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
SIGNIFICANCE Redox-regulated control of protein tyrosine phosphatases (PTPs) through inhibitory reversible oxidation of their active site is emerging as a novel and general mechanism for control of cell surface receptor-activated signaling. This mechanism allows for a previously unrecognized crosstalk between redox regulators and signaling pathways, governed by, for example, receptor tyrosine kinases and integrins, which control cell proliferation and migration. RECENT ADVANCES A large number of different molecules, in addition to hydrogen peroxide, have been found to induce PTP inactivation, including lipid peroxides, reactive nitrogen species, and hydrogen sulfide. Characterization of oxidized PTPs has identified different types of oxidative modifications that are likely to display differential sensitivity to various reducing systems. Accumulating evidence demonstrates that PTP oxidation occurs in a temporally and spatially restricted manner. Studies in cell and animal models indicate altered PTP oxidation in models of common diseases, such as cancer and metabolic/cardiovascular disease. Novel methods have appeared that allow characterization of global PTP oxidation. CRITICAL ISSUES As the understanding of the molecular and cellular biology of PTP oxidation is developing, it will be important to establish experimental procedures that allow analyses of PTP oxidation, and its regulation, in physiological and pathophysiological settings. Future studies should also aim to establish specific connections between various oxidants, specific PTPs, and defined signaling contexts. FUTURE DIRECTIONS Modulation of PTP activity still appears as a valid strategy for correction or inhibition of dys-regulated cell signaling. Continued studies on PTP oxidation might present yet unrecognized means to exploit this regulatory mechanism for pharmacological purposes.
Collapse
Affiliation(s)
- Jeroen Frijhoff
- 1 Department of Oncology-Pathology, Karolinska Institutet , Stockholm, Sweden
| | | | | | | |
Collapse
|
34
|
SUN LI, LI WEIPING, LI WEIZU, XIONG LI, LI GUIPING, MA RONG. Astragaloside IV prevents damage to human mesangial cells through the inhibition of the NADPH oxidase/ROS/Akt/NF-κB pathway under high glucose conditions. Int J Mol Med 2014; 34:167-76. [DOI: 10.3892/ijmm.2014.1741] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 04/01/2014] [Indexed: 11/06/2022] Open
|
35
|
Usatyuk PV, Fu P, Mohan V, Epshtein Y, Jacobson JR, Gomez-Cambronero J, Wary KK, Bindokas V, Dudek SM, Salgia R, Garcia JGN, Natarajan V. Role of c-Met/phosphatidylinositol 3-kinase (PI3k)/Akt signaling in hepatocyte growth factor (HGF)-mediated lamellipodia formation, reactive oxygen species (ROS) generation, and motility of lung endothelial cells. J Biol Chem 2014; 289:13476-91. [PMID: 24634221 DOI: 10.1074/jbc.m113.527556] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hepatocyte growth factor (HGF) mediated signaling promotes cell proliferation and migration in a variety of cell types and plays a key role in tumorigenesis. As cell migration is important to angiogenesis, we characterized HGF-mediated effects on the formation of lamellipodia, a pre-requisite for migration using human lung microvascular endothelial cells (HLMVECs). HGF, in a dose-dependent manner, induced c-Met phosphorylation (Tyr-1234/1235, Tyr-1349, Ser-985, Tyr-1003, and Tyr-1313), activation of PI3k (phospho-Yp85) and Akt (phospho-Thr-308 and phospho-Ser-473) and potentiated lamellipodia formation and HLMVEC migration. Inhibition of c-Met kinase by SU11274 significantly attenuated c-Met, PI3k, and Akt phosphorylation, suppressed lamellipodia formation and endothelial cell migration. LY294002, an inhibitor of PI3k, abolished HGF-induced PI3k (Tyr-458), and Akt (Thr-308 and Ser-473) phosphorylation and suppressed lamellipodia formation. Furthermore, HGF stimulated p47(phox)/Cortactin/Rac1 translocation to lamellipodia and ROS generation. Moreover, inhibition of c-Met/PI3k/Akt signaling axis and NADPH oxidase attenuated HGF- induced lamellipodia formation, ROS generation and cell migration. Ex vivo experiments with mouse aortic rings revealed a role for c-Met signaling in HGF-induced sprouting and lamellipodia formation. Taken together, these data provide evidence in support of a significant role for HGF-induced c-Met/PI3k/Akt signaling and NADPH oxidase activation in lamellipodia formation and motility of lung endothelial cells.
Collapse
|
36
|
HO-1 induction by CO-RM2 attenuates TNF-α-induced cytosolic phospholipase A2 expression via inhibition of PKCα-dependent NADPH oxidase/ROS and NF-κB. Mediators Inflamm 2014; 2014:279171. [PMID: 24616552 PMCID: PMC3927740 DOI: 10.1155/2014/279171] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 11/14/2013] [Accepted: 11/22/2013] [Indexed: 12/27/2022] Open
Abstract
Rheumatoid arthritis (RA) is characterized by chronic inflammatory infiltration of the synovium and elevation of proinflammatory cytokines. Cytosolic phospholipase A2 (cPLA2) is involved in the development of inflammatory diseases. Heme oxygenase-1 (HO-1) has been shown to possess anti-inflammatory properties. The objective of the study was to investigate the detailed mechanisms of TNF-α-induced cPLA2 expression and to determine whether carbon monoxide releasing molecule-2 (CO-RM2) suppresses TNF-α-induced expression of NF-κB-related proinflammatory genes, including cPLA2, via HO-1 induction in RA synovial fibroblasts (RASFs). Here, we reported that TNF-α-induced cPLA2 expression was mediated through TNFR1/PKCα-dependent signaling pathways, including NADPH oxidase (NOX) activation/ROS production and NF-κB activation. CO-RM2 significantly suppressed TNF-α-induced cPLA2 expression by inhibiting the ROS generation and the phosphorylation of NF-κB p65 and IKKα/β, but not the phosphorylation of p38 MAPK and JNK1/2. These results were further confirmed by a ChIP assay to detect the NF-κB DNA-binding activity. Our results demonstrated that induction of HO-1 by CO-RM2 exerted anti-inflammatory and antioxidant effects which were required in concert to prevent the activation of NF-κB leading to induction of various inflammatory genes implicated in the pathogenesis of RA.
Collapse
|
37
|
Wagner B, Gorin Y. Src tyrosine kinase mediates platelet-derived growth factor BB-induced and redox-dependent migration in metanephric mesenchymal cells. Am J Physiol Renal Physiol 2013; 306:F85-97. [PMID: 24197068 DOI: 10.1152/ajprenal.00371.2013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The adult kidney is derived from the interaction between the metanephric blastema and the ureteric bud. Platelet-derived growth factor (PDGF) receptor β is essential for the development of the mature glomerular tuft, as mice deficient for this receptor lack mesangial cells. This study investigated the role of Src tyrosine kinase in PDGF-mediated reactive oxygen species (ROS) generation and migration of metanephric mesenchymal cells (MMCs). Cultured embryonic MMCs from wild-type and PDGF receptor-deficient embryos were established. Migration was determined via wound-healing assay. Unlike PDGF AA, PDGF BB-induced greater migration in MMCs with respect to control. This was abrogated by neutralizing an antibody to PDGF BB. Phosphatidylinositol 3-kinase (PI3K) inhibitors suppressed PDGF BB-induced migration. Conversely, mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK) inhibitors had no effect. Src inhibitors inhibited PDGF-induced cell migration, PI3K activity, and Akt phosphorylation. Adenoviral dominant negative Src (AD DN Src) abrogated PDGF BB-induced Akt phosphorylation. Hydrogen peroxide stimulated cell migration. PDGF BB-induced wound closure was inhibited by the antioxidants N-acetyl-l-cysteine, tiron, and the flavoprotein inhibitor diphenyleneiodonium. These cells express the NADPH oxidase homolog Nox4. Inhibiting Nox4 with antisense oligonucleotides or small interfering RNA (siRNA) suppressed PDGF-induced wound closure. Inhibition of Src with siRNA reduced PDGF BB-induced ROS generation as assessed by 2',7'-dichlorodihydrofluorescein diacetate fluorescence. Furthermore, PDGF BB-stimulated ROS generation and migration were similarly suppressed by Ad DN Src. In MMCs, PDGF BB-induced migration is mediated by PI3K and Src in a redox-dependent manner involving Nox4. Src may be upstream to PI3K and Nox4.
Collapse
Affiliation(s)
- Brent Wagner
- South Texas Veterans Health Care System, Div. of Nephrology MC 7882, 7703 Floyd Curl Dr., San Antonio, TX 78229-3900.
| | | |
Collapse
|
38
|
Guinan AF, Rochfort KD, Fitzpatrick PA, Walsh TG, Pierotti AR, Phelan S, Murphy RP, Cummins PM. Shear stress is a positive regulator of thimet oligopeptidase (EC3.4.24.15) in vascular endothelial cells: consequences for MHC1 levels. Cardiovasc Res 2013; 99:545-54. [DOI: 10.1093/cvr/cvt127] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
39
|
Li H, Peng W, Zhuang J, Lu Y, Jian W, Wei Y, Li W, Xu Y. Vaspin attenuates high glucose-induced vascular smooth muscle cells proliferation and chemokinesis by inhibiting the MAPK, PI3K/Akt, and NF-κB signaling pathways. Atherosclerosis 2013; 228:61-8. [PMID: 23497782 DOI: 10.1016/j.atherosclerosis.2013.02.013] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Revised: 02/06/2013] [Accepted: 02/13/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Vaspin has insulin-sensitizing effects, as well as additional beneficial effects on metabolic diseases. However, little is known about the direct effects of vaspin on vascular complications mediated by diabetes. The objective of this study is to determine the efficacy and mechanism of vaspin on hyperglycemia-induced vascular smooth muscle cells (VSMCs) proliferation, chemokinesis and cell signaling. METHODS Rat VSMCs proliferation was determined with 5-ethynyl-2'-deoxyuridine cell proliferation assays, chemokinesis was monitored with scratch assays, and reactive oxygen species (ROS) production was assessed using H2DCFDA and SOD-inhibited reduction of ferricytochrome c assay. Luciferase activity is assayed using a Dual Luciferase Reporter Assay System. Cell signaling is assessed by immunoblotting. RESULTS Vaspin significantly inhibited VSMCs proliferation and chemokinesis, as well as ROS generation and NADPH oxidase activity, induced by high glucose (HG) treatment. Compared with HG, vaspin significantly decreased VSMCs proliferation by 40 ± 8% at 100 ng/ml. Vaspin also decreased ROS production by 16 ± 8% at 100 ng/ml and 30 ± 8% at 300 ng/ml (all P < 0.01). Vaspin significantly abolished HG-induced phosphorylation of oxidase subunits p47phox, Akt, p38, and JNK1/2 without affecting their total levels, and attenuated HG-induced phosphorylation of insulin receptor and its downstream IRS-1 and IRS-2. For downstream targets, NF-κB activity and IκBα phosphorylation were both enhanced significantly after HG stimulation, and these effects were inhibited by vaspin. Vaspin also significantly abolished HG-induced PCNA and cyclin D1 expression. CONCLUSIONS Vaspin inhibits HG-induced VSMCs proliferation and chemokinesis by preventing ROS activation and MAPK, PI3K/Akt, and NF-κB signaling.
Collapse
Affiliation(s)
- Hailing Li
- Department of Cardiology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, 301 Middle Yanchang Road, Shanghai 200072, China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhao G, Yu R, Deng J, Zhao Q, Li Y, Joo M, van Breemen RB, Christman JW, Xiao L. Pivotal role of reactive oxygen species in differential regulation of lipopolysaccharide-induced prostaglandins production in macrophages. Mol Pharmacol 2013; 83:167-78. [PMID: 23071105 PMCID: PMC3533474 DOI: 10.1124/mol.112.080762] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 10/15/2012] [Indexed: 12/29/2022] Open
Abstract
Gram-negative bacterial endotoxin lipopolysaccharide (LPS) triggers the production of inflammatory cytokines, reactive oxygen species (ROS), and prostaglandins (PGs) by pulmonary macrophages. Here, we investigated if ROS influenced PGs production in response to LPS treatment in mouse bone marrow-derived macrophages (BMDM). We observed that pretreatment of BMDM with two structurally unrelated ROS scavengers, MnTMPyP and EUK-134, not only prevented LPS-induced ROS accumulation, but also attenuated the LPS-induced PGD(2), but not PGE(2), production. Conversely LPS-induced PGD(2), but not PGE(2), production, was potentiated with the cotreatment of BMDM with H(2)O(2). These data suggest that ROS differentially regulate PGD(2) and PGE(2) production in BMDM. In addition, selective inhibition of the ROS generator NADPH oxidase (NOX) using either pharmacologic inhibitors or its p47(phox) subunit deficient mouse BMDM also attenuated LPS-induced PGD(2), but not PGE(2) production, suggesting the critical role of NOX-generated ROS in LPS-induced PGD(2) production in BMDM. We further found that both hematopoietic PGD synthase (H-PGDS) siRNA and its inhibitor HQL-79, but not lipocalin PGDS (L-PGDS) siRNA and its inhibitor AT-56, significantly attenuated LPS-induced PGD(2) production, suggesting that H-PGDS, but not L-PGDS, mediates LPS-induced PGD(2) production in BMDM. Furthermore, data from our in vitro cell-free enzymatic studies showed that coincubation of the recombinant H-PGDS with either MnTMPyP, EUK-134, or catalase significantly decreased PGD(2) production, whereas coincubation with H(2)O(2) significantly increased PGD(2) production. Taken together, our results show that LPS-induced NOX-generated ROS production differentially and specifically regulates the H-PGDS-mediated production of PGD(2), but not PGE(2), in mouse BMDM.
Collapse
Affiliation(s)
- Guiqing Zhao
- Department of Medicine, M/C: 719 University of Illinois at Chicago 840 S. Wood Street Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Lennartsson J, Rönnstrand L. Stem Cell Factor Receptor/c-Kit: From Basic Science to Clinical Implications. Physiol Rev 2012; 92:1619-49. [DOI: 10.1152/physrev.00046.2011] [Citation(s) in RCA: 593] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Stem cell factor (SCF) is a dimeric molecule that exerts its biological functions by binding to and activating the receptor tyrosine kinase c-Kit. Activation of c-Kit leads to its autophosphorylation and initiation of signal transduction. Signaling proteins are recruited to activated c-Kit by certain interaction domains (e.g., SH2 and PTB) that specifically bind to phosphorylated tyrosine residues in the intracellular region of c-Kit. Activation of c-Kit signaling has been found to mediate cell survival, migration, and proliferation depending on the cell type. Signaling from c-Kit is crucial for normal hematopoiesis, pigmentation, fertility, gut movement, and some aspects of the nervous system. Deregulated c-Kit kinase activity has been found in a number of pathological conditions, including cancer and allergy. The observation that gain-of-function mutations in c-Kit can promote tumor formation and progression has stimulated the development of therapeutics agents targeting this receptor, e.g., the clinically used inhibitor imatinib mesylate. Also other clinically used multiselective kinase inhibitors, for instance, sorafenib and sunitinib, have c-Kit included in their range of targets. Furthermore, loss-of-function mutations in c-Kit have been observed and shown to give rise to a condition called piebaldism. This review provides a summary of our current knowledge regarding structural and functional aspects of c-Kit signaling both under normal and pathological conditions, as well as advances in the development of low-molecular-weight molecules inhibiting c-Kit function.
Collapse
Affiliation(s)
- Johan Lennartsson
- Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden; and Experimental Clinical Chemistry, Wallenberg Laboratory, Department of Laboratory Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Lars Rönnstrand
- Ludwig Institute for Cancer Research, Uppsala University, Uppsala, Sweden; and Experimental Clinical Chemistry, Wallenberg Laboratory, Department of Laboratory Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
43
|
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
|
44
|
Ching JK, Luebbert SH, Collins RL, Zhang Z, Marupudi N, Banerjee S, Hurd RD, Ralston L, Fisher JS. Ataxia telangiectasia mutated impacts insulin-like growth factor 1 signalling in skeletal muscle. Exp Physiol 2012; 98:526-35. [PMID: 22941977 DOI: 10.1113/expphysiol.2012.066357] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Reports that ataxia telangiectasia mutated (ATM) is required for full activation of Akt raise the hypothesis that ATM plays a role in insulin-like growth factor 1 (IGF-1) signalling through the Akt/mammalian target of rapamycin (mTOR) pathway. Differentiated C2C12 cells harbouring either ATM-targeting short hairpin RNA (shRNA) or non-targeting shRNA and myotubes from a C2C12 lineage previously exposed to empty vector lentivirus were incubated in the presence or absence of 10 nm IGF-1 followed by Western blot analysis. Parallel experiments were performed in isolated soleus muscles from mice expressing only one functional ATM allele (ATM(+/-)) compared with muscles from wild-type (ATM(+/+)) mice. Insulin-like growth factor 1 increased phosphorylation of Akt S473, Akt T308 and p70 S6 kinase (S6K) in myotubes expressing non-targeting shRNA and in empty vector controls, but the IGF-1 effects were significantly reduced in myotubes with shRNA-mediated ATM knockdown. Likewise, IGF-1-stimulated phosphorylation of Akt S473, Akt T308, mTOR and S6K was lower in isolated soleus muscles from ATM(+/-) mice compared with muscles from ATM(+/+) mice. The ATM inhibitor KU55933 prevented stimulation of S6K phosphorylation in C2C12 myotubes exposed to IGF-1, suggesting that decreased IGF-1 action is not limited to chronic conditions of decreased ATM function. Stimulation of insulin receptor substrate 1 tyrosine 612 phosphorylation by IGF-1 was unaffected by ATM deficiency, though IGF-1 phosphatidylinositol 3-kinase activity tended to be lower in muscle from ATM haploinsufficient mice compared with wild-type muscle. The data suggest that ATM is a modulator of IGF-1 signalling downstream of insulin receptor substrate 1 in skeletal muscle.
Collapse
Affiliation(s)
- James Kain Ching
- Department of Biology, Saint Louis University, St Louis, MO 63103, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Targeting microglia-mediated neurotoxicity: the potential of NOX2 inhibitors. Cell Mol Life Sci 2012; 69:2409-27. [PMID: 22581365 DOI: 10.1007/s00018-012-1015-4] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 04/18/2012] [Accepted: 04/20/2012] [Indexed: 12/24/2022]
Abstract
Microglia are key sentinels of central nervous system health, and their dysfunction has been widely implicated in the progressive nature of neurodegenerative diseases. While microglia can produce a host of factors that are toxic to neighboring neurons, NOX2 has been implicated as a common and essential mechanism of microglia-mediated neurotoxicity. Accumulating evidence indicates that activation of the NOX2 enzyme complex in microglia is neurotoxic, both through the production of extracellular reactive oxygen species that damage neighboring neurons as well as the initiation of redox signaling in microglia that amplifies the pro-inflammatory response. More specifically, evidence supports that NOX2 redox signaling enhances microglial sensitivity to pro-inflammatory stimuli, and amplifies the production of neurotoxic cytokines, to promote chronic and neurotoxic microglial activation. Here, we describe the evidence denoting the role of NOX2 in microglia-mediated neurotoxicity with an emphasis on Alzheimer's and Parkinson's disease, describe available inhibitors that have been tested, and detail evidence of the neuroprotective and therapeutic potential of targeting this enzyme complex to regulate microglia.
Collapse
|
46
|
Reactive oxygen species regulate the levels of dual oxidase (Duox1-2) in human neuroblastoma cells. PLoS One 2012; 7:e34405. [PMID: 22523549 PMCID: PMC3327694 DOI: 10.1371/journal.pone.0034405] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 02/27/2012] [Indexed: 12/03/2022] Open
Abstract
Dual Oxidases (DUOX) 1 and 2 are efficiently expressed in thyroid, gut, lung and immune system. The function and the regulation of these enzymes in mammals are still largely unknown. We report here that DUOX 1 and 2 are expressed in human neuroblastoma SK-N-BE cells as well as in a human oligodendrocyte cell line (MO3-13) and in rat brain and they are induced by platelet derived growth factor (PDGF). The levels of DUOX 1 and 2 proteins and mRNAs are induced by reactive oxygen species (ROS) produced by the membrane NADPH oxidase. As to the mechanism, we find that PDGF stimulates membrane NADPH oxidase to produce ROS, which stabilize DUOX1 and 2 mRNAs and increases the levels of the proteins. Silencing of gp91phox (NOX2), or of the other membrane subunit of NADPH oxidase, p22phox, blocks PDGF induction of DUOX1 and 2. These data unravel a novel mechanism of regulation of DUOX enzymes by ROS and identify a circuitry linking NADPH oxidase activity to DUOX1 and 2 levels in neuroblastoma cells.
Collapse
|
47
|
Liu J, Zhou J, Xing D. Phosphatidylinositol 3-kinase plays a vital role in regulation of rice seed vigor via altering NADPH oxidase activity. PLoS One 2012; 7:e33817. [PMID: 22448275 PMCID: PMC3309022 DOI: 10.1371/journal.pone.0033817] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2011] [Accepted: 02/17/2012] [Indexed: 12/30/2022] Open
Abstract
Phosphatidylinositol 3-kinase (PI3K) has been reported to be important in normal plant growth and stress responses. In this study, it was verified that PI3K played a vital role in rice seed germination through regulating NADPH oxidase activity. Suppression of PI3K activity by inhibitors wortmannin or LY294002 could abate the reactive oxygen species (ROS) formation, which resulted in disturbance to the seed germination. And then, the signal cascades that PI3K promoted the ROS liberation was also evaluated. Diphenylene iodonium (DPI), an NADPH oxidase inhibitor, suppressed most of ROS generation in rice seed germination, which suggested that NADPH oxidase was the main source of ROS in this process. Pharmacological experiment and RT-PCR demonstrated that PI3K promoted the expression of Os rboh9. Moreover, functional analysis by native PAGE and the measurement of the 2, 3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazo-lium-5- carboxanilide (XTT) formazan concentration both showed that PI3K promoted the activity of NADPH oxidase. Furthermore, the western blot analysis of OsRac-1 demonstrated that the translocation of Rac-1 from cytoplasm to plasma membrane, which was known as a key factor in the assembly of NADPH oxidase, was suppressed by treatment with PI3K inhibitors, resulting in the decreased activity of NADPH oxidase. Taken together, these data favored the novel conclusion that PI3K regulated NADPH oxidase activity through modulating the recruitment of Rac-1 to plasma membrane and accelerated the process of rice seed germination.
Collapse
Affiliation(s)
| | | | - Da Xing
- MOE Key Laboratory of Laser Life Science and Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
- * E-mail:
| |
Collapse
|
48
|
Jeong SI, Kim SJ, Kwon TH, Yu KY, Kim SY. Schizandrin prevents damage of murine mesangial cells via blocking NADPH oxidase-induced ROS signaling in high glucose. Food Chem Toxicol 2012; 50:1045-53. [DOI: 10.1016/j.fct.2011.11.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Revised: 11/16/2011] [Accepted: 11/18/2011] [Indexed: 01/16/2023]
|
49
|
Arrizabalaga O, Lacerda HM, Zubiaga AM, Zugaza JL. Rac1 protein regulates glycogen phosphorylase activation and controls interleukin (IL)-2-dependent T cell proliferation. J Biol Chem 2012; 287:11878-90. [PMID: 22337875 DOI: 10.1074/jbc.m111.297804] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Small GTPases of the Rho family have been implicated in important cellular processes such as cell migration and adhesion, protein secretion, and/or gene transcription. In the lymphoid system, these GTPases participate in the signaling cascades that are activated after engagement of antigen receptors. However, little is known about the role that Rho GTPases play in IL-2-mediated responses. Here, we show that IL-2 induces Rac1 activation in Kit 225 T cells. We identified by mass spectrometry the muscle isoform of glycogen phosphorylase (PYGM) as a novel Rac1 effector molecule in IL-2-stimulated cells. The interaction between the active form of Rac1 (Rac1-GTP) and PYGM was established directly through a domain comprising amino acids 191-270 of PYGM that exhibits significant homology with the Rac binding domain of PAK1. The integrity of this region was crucial for PYGM activation. Importantly, IL-2-dependent cellular proliferation was inhibited upon blocking both the activation of Rac1 and the activity of PYGM. These results reveal a new role for Rac1 in cell signaling, showing that this GTPase triggers T cell proliferation upon IL-2 stimulation by associating with PYGM and modulating its enzymatic activity.
Collapse
Affiliation(s)
- Onetsine Arrizabalaga
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country, 48940 Leioa, Spain
| | | | | | | |
Collapse
|
50
|
ten Freyhaus H, Dumitrescu D, Berghausen E, Vantler M, Caglayan E, Rosenkranz S. Imatinib mesylate for the treatment of pulmonary arterial hypertension. Expert Opin Investig Drugs 2011; 21:119-34. [PMID: 22074410 DOI: 10.1517/13543784.2012.632408] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Despite recent advances, pulmonary arterial hypertension (PAH) remains a devastating disease which harbors a poor prognosis. Novel therapeutic approaches directly targeting pulmonary vascular remodeling are warranted. AREAS COVERED This review delineates the current limitations in the management of PAH and focuses on a novel, anti-proliferative therapeutic concept. It will help readers understand the mechanisms of receptor tyrosine kinase signaling, with a special focus on platelet-derived growth factor (PDGF) receptors and their role in the pathobiology of PAH. Furthermore, it provides a comprehensive summary regarding the rationale, efficacy and safety of the tyrosine kinase inhibitor imatinib mesylate , which potently inhibits the PDGF receptor, as an additional treatment option in PAH. EXPERT OPINION PDGF is a potent mitogen for pulmonary vascular smooth muscle cells and represents an important mediator of pulmonary vascular remodeling. Imatinib mesylate, a compound that inhibits the Bcr-Abl kinase and was developed for the treatment of chronic myeloid leukemia, also targets PDGF receptors. Both experimental and clinical data indicate that it reverses the vascular remodeling process even when it is fully established. Results from Phase II and III clinical trials suggest potent and prolonged efficacy in patients with severe PAH (i.e., pulmonary vascular resistance > 800 dynes*s*cm(-5)). Future studies should evaluate the long-term clinical efficacy and safety of imatinib, including patients with less impaired hemodynamics. Based on the current knowledge, this compound is likely to become an additional treatment option for patients with PAH and has the potential to at least partially correct the pathology of the disease.
Collapse
Affiliation(s)
- Henrik ten Freyhaus
- Klinik III für Innere Medizin, Center for Molecular Medicine Cologne, Universität zu Köln, Kerpener Str. 62, 50924 Köln, Germany
| | | | | | | | | | | |
Collapse
|