1
|
Mansour HM, Mohamed AF, Khattab MM, El-Khatib AS. Unveiling the therapeutic prospects of EGFR inhibition in rotenone-mediated parkinsonism in rats: Modulation of dopamine D3 receptor. Brain Res 2024; 1834:148893. [PMID: 38554797 DOI: 10.1016/j.brainres.2024.148893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/01/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons in the substantia nigra. The dopamine D3 receptor (D3R) plays a significant role in the pathogenesis and treatment of PD. Activation of receptor tyrosine kinases (RTKs) inhibits signaling mediated by G protein-coupled receptor (GPCR). Epidermal growth factor receptors (EGFRs) and dopamine D3 receptors in the brain are directly associated with PD, both in terms of its development and potential treatment. Therefore, we investigated the impact of modulating the EGFR, a member of the RTKs family, and the dopamine D3R, a member of the GPCR family. In the present study, 100 mg/kg of lapatinib (LAP) was administered to rotenone-intoxicated rats for three weeks. Our findings indicate that LAP effectively alleviated motor impairment, improved histopathological abnormalities, and restored dopaminergic neurons in the substantia nigra. This restoration was achieved through the upregulation of dopamine D3R and increase of tyrosine hydroxylase (TH) expression, as well as boosting dopamine levels. Furthermore, LAP inhibited the activity of p-EGFR, GRK2, and SCR. Additionally, LAP exhibited antioxidant properties by inhibiting the 4-hydroxynonenal (4-HNE) and PLCγ/PKCβII pathway, while enhancing the antioxidant defense mechanism by increasing GSH-GPX4 pathway. The current study offers insights into the potential repositioning of LAP as a disease-modifying drug for PD. This could be achieved by modulating the dopaminergic system and curbing oxidative stress.
Collapse
Affiliation(s)
- Heba M Mansour
- Central Administration of Biologicals, Innovative Products, and Clinical Studies, Egyptian Drug Authority, EDA, Giza, Egypt
| | - Ahmed F Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Faculty of Pharmacy, King Salman International University (KSIU), South Sinai 46612, Egypt.
| | - Mahmoud M Khattab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Aiman S El-Khatib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
2
|
Shuvalova M, Dmitrieva A, Belousov V, Nosov G. The role of reactive oxygen species in the regulation of the blood-brain barrier. Tissue Barriers 2024:2361202. [PMID: 38808582 DOI: 10.1080/21688370.2024.2361202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
The blood-brain barrier (BBB) regulates the exchange of metabolites and cells between the blood and brain, and maintains central nervous system homeostasis. Various factors affect BBB barrier functions, including reactive oxygen species (ROS). ROS can act as stressors, damaging biological molecules, but they also serve as secondary messengers in intracellular signaling cascades during redox signaling. The impact of ROS on the BBB has been observed in multiple sclerosis, stroke, trauma, and other neurological disorders, making blocking ROS generation a promising therapeutic strategy for BBB dysfunction. However, it is important to consider ROS generation during normal BBB functioning for signaling purposes. This review summarizes data on proteins expressed by BBB cells that can be targets of redox signaling or oxidative stress. It also provides examples of signaling molecules whose impact may cause ROS generation in the BBB, as well as discusses the most common diseases associated with BBB dysfunction and excessive ROS generation, open questions that arise in the study of this problem, and possible ways to overcome them.
Collapse
Affiliation(s)
- Margarita Shuvalova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiia Dmitrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vsevolod Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| | - Georgii Nosov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| |
Collapse
|
3
|
Kim HY, Lee J, Kim HJ, Lee BE, Jeong J, Cho EJ, Jang HJ, Shin KJ, Kim MJ, Chae YC, Lee SE, Myung K, Baik JH, Suh PG, Kim JI. PLCγ1 in dopamine neurons critically regulates striatal dopamine release via VMAT2 and synapsin III. Exp Mol Med 2023; 55:2357-2375. [PMID: 37907739 PMCID: PMC10689754 DOI: 10.1038/s12276-023-01104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 08/05/2023] [Accepted: 08/06/2023] [Indexed: 11/02/2023] Open
Abstract
Dopamine neurons are essential for voluntary movement, reward learning, and motivation, and their dysfunction is closely linked to various psychological and neurodegenerative diseases. Hence, understanding the detailed signaling mechanisms that functionally modulate dopamine neurons is crucial for the development of better therapeutic strategies against dopamine-related disorders. Phospholipase Cγ1 (PLCγ1) is a key enzyme in intracellular signaling that regulates diverse neuronal functions in the brain. It was proposed that PLCγ1 is implicated in the development of dopaminergic neurons, while the physiological function of PLCγ1 remains to be determined. In this study, we investigated the physiological role of PLCγ1, one of the key effector enzymes in intracellular signaling, in regulating dopaminergic function in vivo. We found that cell type-specific deletion of PLCγ1 does not adversely affect the development and cellular morphology of midbrain dopamine neurons but does facilitate dopamine release from dopaminergic axon terminals in the striatum. The enhancement of dopamine release was accompanied by increased colocalization of vesicular monoamine transporter 2 (VMAT2) at dopaminergic axon terminals. Notably, dopamine neuron-specific knockout of PLCγ1 also led to heightened expression and colocalization of synapsin III, which controls the trafficking of synaptic vesicles. Furthermore, the knockdown of VMAT2 and synapsin III in dopamine neurons resulted in a significant attenuation of dopamine release, while this attenuation was less severe in PLCγ1 cKO mice. Our findings suggest that PLCγ1 in dopamine neurons could critically modulate dopamine release at axon terminals by directly or indirectly interacting with synaptic machinery, including VMAT2 and synapsin III.
Collapse
Affiliation(s)
- Hye Yun Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jieun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Hyun-Jin Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Byeong Eun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jaewook Jeong
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Eun Jeong Cho
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Hyun-Jun Jang
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, 58245, Republic of Korea
| | - Kyeong Jin Shin
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Min Ji Kim
- Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Young Chan Chae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Seung Eun Lee
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Ja-Hyun Baik
- Department of Life Sciences, Korea University, Seoul, 02841, Korea
| | - Pann-Ghill Suh
- Korea Brain Research Institute (KBRI), Daegu, 41062, Republic of Korea
| | - Jae-Ick Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea.
| |
Collapse
|
4
|
Merchant JP, Zhu K, Henrion MYR, Zaidi SSA, Lau B, Moein S, Alamprese ML, Pearse RV, Bennett DA, Ertekin-Taner N, Young-Pearse TL, Chang R. Predictive network analysis identifies JMJD6 and other potential key drivers in Alzheimer's disease. Commun Biol 2023; 6:503. [PMID: 37188718 PMCID: PMC10185548 DOI: 10.1038/s42003-023-04791-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Despite decades of genetic studies on late-onset Alzheimer's disease, the underlying molecular mechanisms remain unclear. To better comprehend its complex etiology, we use an integrative approach to build robust predictive (causal) network models using two large human multi-omics datasets. We delineate bulk-tissue gene expression into single cell-type gene expression and integrate clinical and pathologic traits, single nucleotide variation, and deconvoluted gene expression for the construction of cell type-specific predictive network models. Here, we focus on neuron-specific network models and prioritize 19 predicted key drivers modulating Alzheimer's pathology, which we then validate by knockdown in human induced pluripotent stem cell-derived neurons. We find that neuronal knockdown of 10 of the 19 targets significantly modulates levels of amyloid-beta and/or phosphorylated tau peptides, most notably JMJD6. We also confirm our network structure by RNA sequencing in the neurons following knockdown of each of the 10 targets, which additionally predicts that they are upstream regulators of REST and VGF. Our work thus identifies robust neuronal key drivers of the Alzheimer's-associated network state which may represent therapeutic targets with relevance to both amyloid and tau pathology in Alzheimer's disease.
Collapse
Affiliation(s)
- Julie P Merchant
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Neuroscience Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kuixi Zhu
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Marc Y R Henrion
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, Pembroke Place, L3 5QA, UK
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme, PO Box 30096, Blantyre, Malawi
| | - Syed S A Zaidi
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Branden Lau
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
- Arizona Research Labs, Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Sara Moein
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Melissa L Alamprese
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA
| | - Richard V Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Nilüfer Ertekin-Taner
- Department of Neuroscience, Mayo Clinic Florida, Jacksonville, FL, USA
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL, USA
| | - Tracy L Young-Pearse
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Harvard University, Boston, MA, USA.
| | - Rui Chang
- The Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- INTelico Therapeutics LLC, Tucson, AZ, USA.
- PATH Biotech LLC, Tucson, AZ, USA.
| |
Collapse
|
5
|
Yun JH, Lee DH, Jeong HS, Kim SH, Ye SK, Cho CH. STAT3 activation in microglia increases pericyte apoptosis in diabetic retinas through TNF-ɑ/AKT/p70S6 kinase signaling. Biochem Biophys Res Commun 2022; 613:133-139. [PMID: 35561580 DOI: 10.1016/j.bbrc.2022.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/02/2022] [Indexed: 11/19/2022]
Abstract
Diabetic retinopathy (DR) is one of the vascular complications associated with diabetes mellitus. Pericyte loss is an early characteristic phenomenon in DR. However, the mechanism by which pericyte apoptosis occurs in DR is not fully understood. We have focused on the increased STAT3 activation in diabetic retinas because STAT3 activation is associated with inflammation, and persistent chronic inflammation is closely related to retinal lesions. In this study, we demonstrated that STAT3 was activated by IFN-γ and IL-6 that highly expressed in diabetic retinas. We identified TNF-α as a potent inducer of pericyte apoptosis in diabetic retinas from the gene expression analysis and found that STAT3 activation in microglia increased TNF-α expression in the diabetic retinas. We also demonstrated that increased TNF-α expression in microglia caused pericyte apoptosis through downregulating AKT/p70S6 kinase signaling. Moreover, we took advantage of mice lacking STAT3 in microglia and demonstrated that STAT3 ablation in microglia reduced the pericyte apoptosis and TNF-α expression in the diabetic retinas. These results suggest that STAT3 activation in microglia plays an important role in pericyte apoptosis in the diabetic retinas through increased TNF-α expression and provide STAT3 activation in microglia as a potential therapeutic target for preventing pericyte loss in DR.
Collapse
Affiliation(s)
- Jang-Hyuk Yun
- Department of Veterinary Pharmacology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Gangwondo, Republic of Korea
| | - Da-Hye Lee
- Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Han-Seok Jeong
- Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Seung-Hoon Kim
- Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Sang-Kyu Ye
- Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Chung-Hyun Cho
- Department of Biomedical Sciences and Pharmacology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Ischemic/Hypoxic Disease Institute, College of Medicine, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Redox Regulation of Lipid Mobilization in Adipose Tissues. Antioxidants (Basel) 2021; 10:antiox10071090. [PMID: 34356323 PMCID: PMC8301038 DOI: 10.3390/antiox10071090] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/14/2022] Open
Abstract
Lipid mobilization in adipose tissues, which includes lipogenesis and lipolysis, is a paramount process in regulating systemic energy metabolism. Reactive oxygen and nitrogen species (ROS and RNS) are byproducts of cellular metabolism that exert signaling functions in several cellular processes, including lipolysis and lipogenesis. During lipolysis, the adipose tissue generates ROS and RNS and thus requires a robust antioxidant response to maintain tight regulation of redox signaling. This review will discuss the production of ROS and RNS within the adipose tissue, their role in regulating lipolysis and lipogenesis, and the implications of antioxidants on lipid mobilization.
Collapse
|
7
|
Saliakoura M, Rossi Sebastiano M, Pozzato C, Heidel FH, Schnöder TM, Savic Prince S, Bubendorf L, Pinton P, A Schmid R, Baumgartner J, Freigang S, Berezowska SA, Rimessi A, Konstantinidou G. PLCγ1 suppression promotes the adaptation of KRAS-mutant lung adenocarcinomas to hypoxia. Nat Cell Biol 2020; 22:1382-1395. [PMID: 33077911 PMCID: PMC7610419 DOI: 10.1038/s41556-020-00592-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/15/2020] [Indexed: 12/25/2022]
Abstract
Mutant KRAS modulates the metabolic plasticity of cancer cells conferring growth advantage during hypoxia, but the molecular underpinnings are largely unknown. Using a lipidomic screen, we found that PLCγ1 is suppressed during hypoxia in KRAS-mutant human lung adenocarcinoma cancer cell lines. Suppression of PLCγ1 in hypoxia promotes a less oxidative cancer cell metabolism, reduces the formation of mitochondrial reactive oxygen species and switches tumor bioenergetics towards glycolysis by impairing Ca2+ entry into the mitochondria. This event prevents lipid peroxidation, antagonizes apoptosis and increases cancer cell proliferation. Accordingly, loss-of-function of Plcγ1 in a mouse model of KrasG12D-driven lung adenocarcinoma increased the expression of glycolytic genes, boosted tumor growth and reduced survival. In patients with mutant KRAS lung adenocarcinomas, low PLCγ1 expression correlates with increased expression of hypoxia markers and predicts poor patient survival. Thus, our work reveals a mechanism of cancer cell adaptation to hypoxia with potential therapeutic value.
Collapse
Affiliation(s)
- Maria Saliakoura
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | | - Chiara Pozzato
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Florian H Heidel
- Internal Medicine II, Hematology and Oncology, University Hospital Jena, Jena, Germany.,Leibniz-Institute on Aging, Fritz-Lipmann-Institute, Jena, Jena, Germany
| | - Tina M Schnöder
- Internal Medicine II, Hematology and Oncology, University Hospital Jena, Jena, Germany.,Leibniz-Institute on Aging, Fritz-Lipmann-Institute, Jena, Jena, Germany
| | | | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Paolo Pinton
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Ralph A Schmid
- Department of General Thoracic Surgery, Inselspital, Bern, Switzerland
| | | | - Stefan Freigang
- Institute of Pathology, University of Bern, Bern, Switzerland
| | | | - Alessandro Rimessi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | | |
Collapse
|
8
|
The Influence of Reactive Oxygen Species in the Immune System and Pathogenesis of Multiple Sclerosis. Autoimmune Dis 2020; 2020:5793817. [PMID: 32789026 PMCID: PMC7334772 DOI: 10.1155/2020/5793817] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 04/14/2020] [Accepted: 05/22/2020] [Indexed: 02/08/2023] Open
Abstract
Multiple roles have been indicated for reactive oxygen species (ROS) in the immune system in recent years. ROS have been extensively studied due to their ability to damage DNA and other subcellular structures. Noticeably, they have been identified as a pivotal second messenger for T-cell receptor signaling and T-cell activation and participate in antigen cross-presentation and chemotaxis. As an agent with direct toxic effects on cells, ROS lead to the initiation of the autoimmune response. Moreover, ROS levels are regulated by antioxidant systems, which include enzymatic and nonenzymatic antioxidants. Enzymatic antioxidants include superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase. Nonenzymatic antioxidants contain vitamins C, A, and E, glutathione, and thioredoxin. Particularly, cellular antioxidant systems have important functions in maintaining the redox system homeostasis. This review will discuss the significant roles of ROS generation and antioxidant systems under normal conditions, in the immune system, and pathogenesis of multiple sclerosis.
Collapse
|
9
|
Kim MJ, Choi WG, Ahn KJ, Chae IG, Yu R, Back SH. Reduced EGFR Level in eIF2α PhosphorylationDeficient Hepatocytes Is Responsible for Susceptibility to Oxidative Stress. Mol Cells 2020; 43:264-275. [PMID: 32150794 PMCID: PMC7103887 DOI: 10.14348/molcells.2020.2197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 12/17/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species (ROS) play a significant role in intracellular signaling and regulation, particularly when they are maintained at physiologic levels. However, excess ROS can cause cell damage and induce cell death. We recently reported that eIF2α phosphorylation protects hepatocytes from oxidative stress and liver fibrosis induced by fructose metabolism. Here, we found that hepatocyte-specific eIF2α phosphorylation-deficient mice have significantly reduced expression of the epidermal growth factor receptor (EGFR) and altered EGFR-mediated signaling pathways. EGFR-mediated signaling pathways are important for cell proliferation, differentiation, and survival in many tissues and cell types. Therefore, we studied whether the reduced amount of EGFR is responsible for the eIF2α phosphorylationdeficient hepatocytes' vulnerability to oxidative stress. ROS such as hydrogen peroxide and superoxides induce both EGFR tyrosine phosphorylation and eIF2α phosphorylation. eIF2α phosphorylation-deficient primary hepatocytes, or EGFR knockdown cells, have decreased ROS scavenging ability compared to normal cells. Therefore, these cells are particularly susceptible to oxidative stress. However, overexpression of EGFR in these eIF2α phosphorylationdeficient primary hepatocytes increased ROS scavenging ability and alleviated ROS-mediated cell death. Therefore, we hypothesize that the reduced EGFR level in eIF2α phosphorylation-deficient hepatocytes is one of critical factors responsible for their susceptibility to oxidative stress.
Collapse
Affiliation(s)
- Mi-Jeong Kim
- School of Biological Sciences, University of Ulsan, Ulsan 4460, Korea
| | - Woo-Gyun Choi
- School of Biological Sciences, University of Ulsan, Ulsan 4460, Korea
| | - Kyung-Ju Ahn
- School of Biological Sciences, University of Ulsan, Ulsan 4460, Korea
| | - In Gyeong Chae
- School of Biological Sciences, University of Ulsan, Ulsan 4460, Korea
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan 44610, Korea
| | - Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan 4460, Korea
| |
Collapse
|
10
|
Fukuyama T, Nakamura Y, Kanemaru K, Toyoda C, Jang HJ, Suh PG, Fukami K. Phospholipase Cγ1 is required for normal irritant contact dermatitis responses and sebaceous gland homeostasis. Exp Dermatol 2019; 28:1051-1057. [PMID: 31338881 DOI: 10.1111/exd.14009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/14/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022]
Abstract
Differentiation and proliferation of keratinocyte are controlled by various signalling pathways. The epidermal growth factor receptor (EGFR) is known to be an important regulator of multiple epidermal functions. Inhibition of EGFR signalling disturbs keratinocyte proliferation, differentiation and migration. Previous studies have revealed that one of the EGFR downstream signalling molecules, phospholipase Cγ1 (PLCγ1), regulates differentiation, proliferation and migration of keratinocytes in in vitro cell culture system. However, the role of PLCγ1 in the regulation of keratinocyte functions in animal epidermis remains unexplored. In this study, we generated keratinocyte-specific PLCγ1 knockout (KO) mice (PLCγ1 cKO mice). Contrary to our expectations, loss of PLCγ1 did not affect differentiation, proliferation and migration of interfollicular keratinocytes. We further examined the role of PLCγ1 in irritant contact dermatitis (ICD), in which epidermal cells play a pivotal role. Upon irritant stimulation, PLCγ1 cKO mice showed exaggerated ICD responses. Further study revealed that epidermal loss of PLCγ1 induced sebaceous gland hyperplasia, indicating that PLCγ1 regulates homeostasis of one of the epidermal appendages. Taken together, our results indicate that, although PLCγ1 is dispensable in interfollicular keratinocyte for normal differentiation, proliferation and migration, it is required for normal ICD responses. Our results also indicate that PLCγ1 regulates homeostasis of sebaceous glands.
Collapse
Affiliation(s)
- Takatsugu Fukuyama
- Laboratory of Genome and Biosignals, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yoshikazu Nakamura
- Laboratory of Genome and Biosignals, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.,PRIME, Japan Agency for Medical Research and Development, Tokyo, Japan.,Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Kaori Kanemaru
- Laboratory of Genome and Biosignals, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.,Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Chiho Toyoda
- Laboratory of Genome and Biosignals, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hyun-Jun Jang
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Pann-Ghill Suh
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Korea.,Korea Brain Reaseach Institute, Daegu, Korea
| | - Kiyoko Fukami
- Laboratory of Genome and Biosignals, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan.,AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
| |
Collapse
|
11
|
Islas-Robles A, Yedlapudi D, Lau SS, Monks TJ. Toxicoproteomic Analysis of Poly(ADP-ribose)-associated Proteins Induced by Oxidative Stress in Human Proximal Tubule Cells. Toxicol Sci 2019; 171:117-131. [PMID: 31165168 DOI: 10.1093/toxsci/kfz131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/28/2019] [Accepted: 05/26/2019] [Indexed: 11/14/2022] Open
Abstract
2,3,5-Tris-(glutathion-S-yl)hydroquinone (TGHQ) is a nephrotoxic and nephrocarcinogenic metabolite of hydroquinone. TGHQ generates ROS, causing DNA strand breaks, hyperactivation of PARP-1, increases in intracellular calcium ([Ca2+]i), and cell death. PARP-1 catalyzes the attachment of ADP-ribose polymers (PAR) to target proteins. In human kidney proximal tubule cells (HK-2), ROS-mediated PARP-1 hyperactivation and elevations in [Ca2+]i are reciprocally coupled. The molecular mechanism of this interaction is unclear. The aim of the present study was to identify ROS-induced PAR-associated proteins to further understand their potential role in cell death. PAR-associated proteins were enriched by immunoprecipitation, identified by LC-MS/MS, and relative abundance was obtained by spectral counting. 356 proteins were PAR-modified following TGHQ treatment. 13 proteins exhibited gene ontology annotations related to calcium. Among these proteins, the general transcription factor II-I (TFII-I) is directly involved in the modulation of [Ca2+]i. TFII-I binding to phospholipase C (PLC) leads to calcium influx via the TRPC3 channel. However, inhibition of TRPC3 or PLC had no effect on TGHQ-mediated cell death, suggesting that their loss of function may be necessary but insufficient to cause cell death. Nevertheless, TGHQ promoted a time-dependent translocation of TFII-I from the nucleus to the cytosol concomitant with a decrease in tyrosine phosphorylation in α/β-TFII-I. Therefore it is likely that ROS have an important impact on the function of TFII-I, such as regulation of transcription, and DNA translesion synthesis. Our data also sheds light on PAR mediated signaling during oxidative stress, and contributes to the development of strategies to prevent PAR-dependent cell death.
Collapse
Affiliation(s)
- Argel Islas-Robles
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721.,Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, Wayne State University, MI 48201
| | - Deepthi Yedlapudi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, Wayne State University, MI 48201
| | - Serrine S Lau
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, Wayne State University, MI 48201
| | - Terrence J Monks
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Detroit, Wayne State University, MI 48201
| |
Collapse
|
12
|
Che D, Hou Y, Zeng Y, Li C, Zhang Y, Wei D, Hu S, Liu R, An H, Wang Y, Zhang T. Dehydroandrographolide inhibits IgE-mediated anaphylactic reactions via calcium signaling pathway. Toxicol Appl Pharmacol 2019; 366:46-53. [DOI: 10.1016/j.taap.2019.01.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 01/16/2019] [Accepted: 01/22/2019] [Indexed: 01/23/2023]
|
13
|
Socodato R, Portugal CC, Rodrigues A, Henriques J, Rodrigues C, Figueira C, Relvas JB. Redox tuning of Ca 2+ signaling in microglia drives glutamate release during hypoxia. Free Radic Biol Med 2018; 118:137-149. [PMID: 29501565 DOI: 10.1016/j.freeradbiomed.2018.02.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/31/2018] [Accepted: 02/27/2018] [Indexed: 02/07/2023]
Abstract
Hypoxia causes oxidative stress and excitotoxicity, culminating in neuronal damage during brain ischemia. Hypoxia also activates microglia, the myeloid resident cells of the brain parenchyma. Upon activation, microglia release high amounts of the neurotransmitter glutamate, contributing for neuronal excitotoxicity during brain insults. Here, we reveal a signaling pathway controlling glutamate release from human microglia during hypoxia. We show that hypoxia-mediated redox imbalance promotes the activation of endoplasmic reticulum inositol 1,4,5-trisphosphate (InsP3) receptors leading to Ca2+ mobilization into the cytosol. Increasing cytosolic Ca2+ signaling in microglia activates the non-receptor protein tyrosine kinase Src at the plasma membrane. Src activation enhances the permeability of microglial gap junctions promoting the release of glutamate during hypoxia. Preventing the hypoxia-triggered redox imbalance, using the dietary antioxidants neochlorogenic acid or vitamin C, inhibits InsP3-dependent Ca2+ signaling and abrogates the release of glutamate. Overall, modulating microglial Ca2+ signaling in response to changes in the redox microenvironment might be critical for controlling glutamate excitotoxicity during hypoxia.
Collapse
Affiliation(s)
- Renato Socodato
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal.
| | - Camila C Portugal
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal
| | - Artur Rodrigues
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal
| | - Joana Henriques
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal
| | - Carla Rodrigues
- Diverge, Grupo Nabeiro Innovation Center, Alameda dos Oceanos 65, 1.1, 1900-208 Lisbon, Portugal
| | - Cláudia Figueira
- Diverge, Grupo Nabeiro Innovation Center, Alameda dos Oceanos 65, 1.1, 1900-208 Lisbon, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde and Instituto de Biologia Molecular e Celular (IBMC) and The Discoveries Centre for Regeneration and Precision Medicine - Porto campus, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
14
|
Yadav VR, Song T, Mei L, Joseph L, Zheng YM, Wang YX. PLCγ1-PKCε-IP 3R1 signaling plays an important role in hypoxia-induced calcium response in pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2018; 314:L724-L735. [PMID: 29388468 DOI: 10.1152/ajplung.00243.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hypoxia-induced pulmonary vasoconstriction (HPV) is attributed to an increase in intracellular Ca2+ concentration ([Ca2+]i) in pulmonary artery smooth muscle cells (PASMCs). We have reported that phospholipase C-γ1 (PLCγ1) plays a significant role in the hypoxia-induced increase in [Ca2+]i in PASMCs and attendant HPV. In this study, we intended to determine molecular mechanisms for hypoxic Ca2+ and contractile responses in PASMCs. Our data reveal that hypoxic vasoconstriction occurs in pulmonary arteries, but not in mesenteric arteries. Hypoxia caused a large increase in [Ca2+]i in PASMCs, which is diminished by the PLC inhibitor U73122 and not by its inactive analog U73433 . Hypoxia augments PLCγ1-dependent inositol 1,4,5-trisphosphate (IP3) generation. Exogenous ROS, hydrogen peroxide (H2O2), increases PLCγ1 phosphorylation at tyrosine-783 and IP3 production. IP3 receptor-1 (IP3R1) knock-down remarkably diminishes hypoxia- or H2O2-induced increase in [Ca2+]i. Hypoxia or H2O2 increases the activity of IP3Rs, which is significantly reduced in protein kinase C-ε (PKCε) knockout PASMCs. A higher PLCγ1 expression, activity, and basal [Ca2+]i are found in PASMCs, but not in mesenteric artery smooth muscle cells from mice exposed to chronic hypoxia (CH) for 21 days. CH enhances H2O2- and ATP-induced increase in [Ca2+]i in PASMCs and PLC-dependent, norepinephrine-evoked pulmonary vasoconstriction. In conclusion, acute hypoxia uniquely causes ROS-dependent PLCγ1 activation, IP3 production, PKCε activation, IP3R1 opening, Ca2+ release, and contraction in mouse PASMCs; CH enhances PASM PLCγ1 expression, activity, and function, playing an essential role in pulmonary hypertension in mice.
Collapse
Affiliation(s)
- Vishal R Yadav
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Tengyao Song
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Lin Mei
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Leroy Joseph
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College , Albany, New York
| |
Collapse
|
15
|
Lopez-Guerrero AM, Pascual-Caro C, Martin-Romero FJ, Pozo-Guisado E. Store-operated calcium entry is dispensable for the activation of ERK1/2 pathway in prostate cancer cells. Cell Signal 2017; 40:44-52. [DOI: 10.1016/j.cellsig.2017.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/23/2017] [Accepted: 08/28/2017] [Indexed: 01/10/2023]
|
16
|
Sreedhar A, Lefort J, Petruska P, Gu X, Shi R, Miriyala S, Panchatcharam M, Zhao Y. UCP2 upregulation promotes PLCγ-1 signaling during skin cell transformation. Mol Carcinog 2017; 56:2290-2300. [PMID: 28574619 PMCID: PMC5582995 DOI: 10.1002/mc.22684] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/22/2017] [Accepted: 06/01/2017] [Indexed: 12/15/2022]
Abstract
Uncoupling protein 2 (UCP2), whose physiological role is to decrease mitochondrial membrane potential and reactive oxygen species (ROS) production, is often overexpressed in human cancers. UCP2 upregulation has recently been proposed as a novel survival mechanism for cancer cells. However, until now, how exactly UCP2 promotes tumorigenesis remains inconclusive. Based on a widely used skin cell transformation model, our data demonstrated that UCP2 differentially regulated ROS. UCP2 upregulation decreased superoxide whereas it increased hydrogen peroxide production with concomitant increase in the expression and activity of manganese superoxide dismutase (MnSOD), the primary mitochondrial antioxidant enzyme. Furthermore, hydrogen peroxide was responsible for induction of lipid peroxidation, and PLCγ-1 activation in UCP2 overexpressed cells. Additionally, PLCγ-1 activation enhanced skin cell transformation, and pharmacological, and siRNA mediated inhibition of PLCγ-1, markedly reduced colony formation, and 3D cell growth. Moreover, hydrogen peroxide scavenger, catalase, suppressed lipid peroxidation, and dampened PLCγ-1 activity. Taken together, our data suggest that (i) UCP2 is an important regulator of mitochondrial redox status and lipid signaling; (ii) hydrogen peroxide might mediate UCP2's tumor promoting activity; and (iii) pharmacological disruption of PLCγ-1 and/or hydrogen peroxide may have clinical utility for UCP2 overexpressed cancers.
Collapse
Affiliation(s)
- Annapoorna Sreedhar
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA
| | - Julia Lefort
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA
| | - Petra Petruska
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA
| | - Xin Gu
- Department of Pathology, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA
| | - Runhua Shi
- Feist-Weiller Cancer Center, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA
| | - Sumitra Miriyala
- Department of Anatomy and Cell Biology, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA
| | - Manikandan Panchatcharam
- Department of Anatomy and Cell Biology, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA
| | - Yunfeng Zhao
- Department of Pharmacology, Toxicology & Neuroscience, LSU Health Sciences Center in Shreveport, Shreveport, LA 71130, USA
| |
Collapse
|
17
|
Emmanouilidi A, Lattanzio R, Sala G, Piantelli M, Falasca M. The role of phospholipase Cγ1 in breast cancer and its clinical significance. Future Oncol 2017; 13:1991-1997. [DOI: 10.2217/fon-2017-0125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Breast cancer, the most common malignancy among women, is usually detected at an early stage and has a low risk of relapse. Nevertheless, a significant number of patients cannot be cured solely by local treatment. Distinguishing between patients who are of low risk of relapse from those who are of high risk may have important implications to improve treatment outcomes. The PLC-γ1 signaling pathway promotes many physiological processes, including cell migration and invasion. Increasing evidence shows aberrant PLC-γ1 signaling implication in carcinogenesis including breast cancer. In this review, the role of PLC-γ1 in breast cancer and its clinical implications will be discussed, as well as its potential as a prognostic factor and a therapeutic target.
Collapse
Affiliation(s)
- Aikaterini Emmanouilidi
- Curtin Health Innovation Research Institute, School of Biomedical Sciences, Curtin University, Perth, Western Australia, Australia
| | - Rossano Lattanzio
- Department of Medical, Oral & Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Gianluca Sala
- Department of Medical, Oral & Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Mauro Piantelli
- Department of Medical, Oral & Biotechnological Sciences, G. d'Annunzio University, Chieti, Italy
| | - Marco Falasca
- Curtin Health Innovation Research Institute, School of Biomedical Sciences, Curtin University, Perth, Western Australia, Australia
| |
Collapse
|
18
|
Xu Q, Huff LP, Fujii M, Griendling KK. Redox regulation of the actin cytoskeleton and its role in the vascular system. Free Radic Biol Med 2017; 109:84-107. [PMID: 28285002 PMCID: PMC5497502 DOI: 10.1016/j.freeradbiomed.2017.03.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 02/17/2017] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
The actin cytoskeleton is critical for form and function of vascular cells, serving mechanical, organizational and signaling roles. Because many cytoskeletal proteins are sensitive to reactive oxygen species, redox regulation has emerged as a pivotal modulator of the actin cytoskeleton and its associated proteins. Here, we summarize work implicating oxidants in altering actin cytoskeletal proteins and focus on how these alterations affect cell migration, proliferation and contraction of vascular cells. Finally, we discuss the role of oxidative modification of the actin cytoskeleton in vivo and highlight its importance for vascular diseases.
Collapse
Affiliation(s)
- Qian Xu
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States; Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Lauren P Huff
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States
| | - Masakazu Fujii
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Japan
| | - Kathy K Griendling
- Division of Cardiology, Department of Medicine, Emory University, 101 Woodruff Circle, 308a WMB, Atlanta, GA 30322, United States.
| |
Collapse
|
19
|
Decrock E, Hoorelbeke D, Ramadan R, Delvaeye T, De Bock M, Wang N, Krysko DV, Baatout S, Bultynck G, Aerts A, Vinken M, Leybaert L. Calcium, oxidative stress and connexin channels, a harmonious orchestra directing the response to radiotherapy treatment? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1099-1120. [DOI: 10.1016/j.bbamcr.2017.02.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 02/02/2017] [Accepted: 02/04/2017] [Indexed: 02/07/2023]
|
20
|
Sulfite triggers sustained calcium overload in cultured cortical neurons via a redox-dependent mechanism. Toxicol Lett 2016; 258:237-248. [DOI: 10.1016/j.toxlet.2016.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/10/2016] [Accepted: 06/12/2016] [Indexed: 12/25/2022]
|
21
|
Li C, Tong H, Yan Q, Tang S, Han X, Xiao W, Tan Z. L-Theanine Improves Immunity by Altering TH2/TH1 Cytokine Balance, Brain Neurotransmitters, and Expression of Phospholipase C in Rat Hearts. Med Sci Monit 2016; 22:662-9. [PMID: 26922362 PMCID: PMC4772912 DOI: 10.12659/msm.897077] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Background This study aimed to investigate the regulatory effects of L-theanine on secretion of immune cytokines, hormones, and neurotransmitters, and mRNA expression of phospholipase C (PLC) in rats, and to explore its regulatory mechanism in immune function. Material/Methods Sixty-four Sprague-Dawley rats received daily intragastric infusion of different doses of L-theanine solution [0, 50 (LT), 200 (MT), and 400 (HT) mg/kg BW]. Cytokines, immunoglobulins, and hormones in the serum, neurotransmitters, and mRNA expression of PLC in the relevant tissues were assayed. Results L-theanine administration increased the splenic organ index and decreased the contents of ILs-4/6/10 and the ratio of IL-4/IFN-γ in the serum. High-dose L-theanine administration increased the levels of dopamine and 5-hydroxytryptamine in the pituitary and hippocampus, resulting in decrease in corticosterone level in the serum. L-theanine administration decreased the mRNA expressions of PLC isomers in the liver and PLC-γ1 and PLC-δ1 in the spleen. Interestingly, mRNA expressions of PLC-βf1 in the spleen and PLC isomers mRNA in the heart were up-regulated by L-theanine administration. Conclusions Administration of 400 mg/kg BWL-theanine improved immune function of the rats by increasing the splenic weight, altering the Th2/Th1 cytokine balance, decreasing the corticosterone level in the serum, elevating dopamine and 5-hydroxytryptamine in the brain, and regulating the mRNA expression of PLC isomers in the heart.
Collapse
Affiliation(s)
- Chengjian Li
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients from Botanicals, Provincial Co-Innovation Center for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan, China (mainland)
| | - Haiou Tong
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients from Botanicals, Provincial Co-Innovation Center for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan, China (mainland)
| | - Qiongxian Yan
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China (mainland)
| | - Shaoxun Tang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Research Center of Livestock & Poultry Sciences, South-Central Experimental Station of Animal Nutrition and Feed Science in Ministry of Agriculture, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, Hunan, China (mainland)
| | - Xuefeng Han
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients from Botanicals, Provincial Co-Innovation Center for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan, China (mainland)
| | - Wenjun Xiao
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients from Botanicals, Provincial Co-Innovation Center for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan, China (mainland)
| | - Zhiliang Tan
- National Research Center of Engineering Technology for Utilization of Botanical Functional Ingredients from Botanicals, Provincial Co-Innovation Center for Utilization of Botanical Function Ingredients, Hunan Agricultural University, Changsha, Hunan, China (mainland)
| |
Collapse
|
22
|
Gajski G, Domijan AM, Žegura B, Štern A, Gerić M, Novak Jovanović I, Vrhovac I, Madunić J, Breljak D, Filipič M, Garaj-Vrhovac V. Melittin induced cytogenetic damage, oxidative stress and changes in gene expression in human peripheral blood lymphocytes. Toxicon 2016; 110:56-67. [DOI: 10.1016/j.toxicon.2015.12.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 12/07/2015] [Accepted: 12/11/2015] [Indexed: 12/12/2022]
|
23
|
Hiramatsu N, Chiang WC, Kurt TD, Sigurdson CJ, Lin JH. Multiple Mechanisms of Unfolded Protein Response-Induced Cell Death. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:1800-8. [PMID: 25956028 PMCID: PMC4484218 DOI: 10.1016/j.ajpath.2015.03.009] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 02/09/2015] [Accepted: 03/26/2015] [Indexed: 12/12/2022]
Abstract
Eukaryotic cells fold and assemble membrane and secreted proteins in the endoplasmic reticulum (ER), before delivery to other cellular compartments or the extracellular environment. Correctly folded proteins are released from the ER, and poorly folded proteins are retained until they achieve stable conformations; irreparably misfolded proteins are targeted for degradation. Diverse pathological insults, such as amino acid mutations, hypoxia, or infection, can overwhelm ER protein quality control, leading to misfolded protein buildup, causing ER stress. To cope with ER stress, eukaryotic cells activate the unfolded protein response (UPR) by increasing levels of ER protein-folding enzymes and chaperones, enhancing the degradation of misfolded proteins, and reducing protein translation. In mammalian cells, three ER transmembrane proteins, inositol-requiring enzyme-1 (IRE1; official name ERN1), PKR-like ER kinase (PERK; official name EIF2AK3), and activating transcription factor-6, control the UPR. The UPR signaling triggers a set of prodeath programs when the cells fail to successfully adapt to ER stress or restore homeostasis. ER stress and UPR signaling are implicated in the pathogenesis of diverse diseases, including neurodegeneration, cancer, diabetes, and inflammation. This review discusses the current understanding in both adaptive and apoptotic responses as well as the molecular mechanisms instigating apoptosis via IRE1 and PERK signaling. We also examine how IRE1 and PERK signaling may be differentially used during neurodegeneration arising in retinitis pigmentosa and prion infection.
Collapse
Affiliation(s)
- Nobuhiko Hiramatsu
- Department of Pathology, University of California-San Diego, La Jolla, California
| | - Wei-Chieh Chiang
- Department of Pathology, University of California-San Diego, La Jolla, California
| | - Timothy D Kurt
- Department of Pathology, University of California-San Diego, La Jolla, California
| | | | - Jonathan H Lin
- Department of Pathology, University of California-San Diego, La Jolla, California.
| |
Collapse
|
24
|
Steinberg SF. Mechanisms for redox-regulation of protein kinase C. Front Pharmacol 2015; 6:128. [PMID: 26157389 PMCID: PMC4477140 DOI: 10.3389/fphar.2015.00128] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 06/10/2015] [Indexed: 11/21/2022] Open
Abstract
Protein kinase C (PKC) is comprised of a family of signal-regulated enzymes that play pleiotropic roles in the control of many physiological and pathological responses. PKC isoforms are traditionally viewed as allosterically activated enzymes that are recruited to membranes by growth factor receptor-generated lipid cofactors. An inherent assumption of this conventional model of PKC isoform activation is that PKCs act exclusively at membrane-delimited substrates and that PKC catalytic activity is an inherent property of each enzyme that is not altered by the activation process. This traditional model of PKC activation does not adequately explain the many well-documented actions of PKC enzymes in mitochondrial, nuclear, and cardiac sarcomeric (non-sarcolemmal) subcellular compartments. Recent studies address this dilemma by identifying stimulus-specific differences in the mechanisms for PKC isoform activation during growth factor activation versus oxidative stress. This review discusses a number of non-canonical redox-triggered mechanisms that can alter the catalytic properties and subcellular compartmentation patterns of PKC enzymes. While some redox-activated mechanisms act at structural determinants that are common to all PKCs, the redox-dependent mechanism for PKCδ activation requires Src-dependent tyrosine phosphorylation of a unique phosphorylation motif on this enzyme and is isoform specific. Since oxidative stress contributes to pathogenesis of a wide range of clinical disorders, these stimulus-specific differences in the controls and consequences of PKC activation have important implications for the design and evaluation of PKC-targeted therapeutics.
Collapse
Affiliation(s)
- Susan F Steinberg
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University New York, NY, USA
| |
Collapse
|
25
|
Piazzi M, Blalock WL, Bavelloni A, Faenza I, Raffini M, Tagliavini F, Manzoli L, Cocco L. PI-PLCβ1b affects Akt activation, cyclin E expression, and caspase cleavage, promoting cell survival in pro-B-lymphoblastic cells exposed to oxidative stress. FASEB J 2014; 29:1383-94. [PMID: 25550457 DOI: 10.1096/fj.14-259051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 12/01/2014] [Indexed: 12/31/2022]
Abstract
The phosphoinositide-dependent signal transduction pathway has been implicated in the control of a variety of biologic processes, such as the regulation of cellular metabolism and homeostasis, cell proliferation and differentiation, and apoptosis. One of the key players in the regulation of inositol lipid signaling is the phospholipase Cβ1 (PI-PLCβ1), that hydrolyzes phosphatidylinositol 4,5-bisphosphate [PtIns(4,5)P2], giving rise to the second messengers inositol triphosphate and diacylglicerol. PI-PLCβ1 has been associated with the regulation of several cellular functions, some of which have not yet been fully understood. In particular, it has been reported that PI-PLCβ1 protects murine fibroblasts from oxidative stress-induced cell death. The mediators of oxidative stress, reactive oxygen species (ROS), have been shown to regulate major epigenetic processes, causing the silencing of tumor suppressors and enhancing the proliferation of leukemic cells under oxidative stress. Investigation of the interplay between ROS, PI-PLCβ1, and their signaling mediators in leukemia might therefore reveal innovative targets of pharmacological therapy in the treatment for leukemia. In this work, we demonstrate that in pro-B-lymphoblastic cells (Ba/F3), treated with H2O2, PI-PLCβ1b conferred resistance to cell death, promoting cell cycle progression and cell proliferation and influencing the expression of cyclin A and E. Interestingly, we found that, expression of PI-PLCβ1b affects the activity of caspase-3, caspase-7, and of several protein kinases induced by oxidative stress. In particular, PI-PLCβ1b expression completely abolished the phosphorylation of Erk1/2 MAP kinases, down-regulated phosphatase and tensin homolog (PTEN), and up-regulated the phosphorylation of Akt, thereby sustaining cellular proliferation.
Collapse
Affiliation(s)
- Manuela Piazzi
- *Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy; Struttura Complessa Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy; Institute of Molecular Genetics, National Research Council of Italy, Bologna, Italy; and RAMSES Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - William L Blalock
- *Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy; Struttura Complessa Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy; Institute of Molecular Genetics, National Research Council of Italy, Bologna, Italy; and RAMSES Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Alberto Bavelloni
- *Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy; Struttura Complessa Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy; Institute of Molecular Genetics, National Research Council of Italy, Bologna, Italy; and RAMSES Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Irene Faenza
- *Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy; Struttura Complessa Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy; Institute of Molecular Genetics, National Research Council of Italy, Bologna, Italy; and RAMSES Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Mirco Raffini
- *Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy; Struttura Complessa Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy; Institute of Molecular Genetics, National Research Council of Italy, Bologna, Italy; and RAMSES Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Francesca Tagliavini
- *Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy; Struttura Complessa Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy; Institute of Molecular Genetics, National Research Council of Italy, Bologna, Italy; and RAMSES Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Lucia Manzoli
- *Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy; Struttura Complessa Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy; Institute of Molecular Genetics, National Research Council of Italy, Bologna, Italy; and RAMSES Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| | - Lucio Cocco
- *Cell Signaling Laboratory, Department of Biomedical Sciences, University of Bologna, Bologna, Italy; Struttura Complessa Laboratory of Musculoskeletal Cell Biology, Rizzoli Orthopedic Institute, Bologna, Italy; Institute of Molecular Genetics, National Research Council of Italy, Bologna, Italy; and RAMSES Laboratory, Rizzoli Orthopedic Institute, Bologna, Italy
| |
Collapse
|
26
|
Kunze K, Spieker T, Gamerdinger U, Nau K, Berger J, Dreyer T, Sindermann JR, Hoffmeier A, Gattenlöhner S, Bräuninger A. A recurrent activating PLCG1 mutation in cardiac angiosarcomas increases apoptosis resistance and invasiveness of endothelial cells. Cancer Res 2014; 74:6173-83. [PMID: 25252913 DOI: 10.1158/0008-5472.can-14-1162] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Primary cardiac angiosarcomas are rare tumors with unfavorable prognosis. Pathogenic driver mutations are largely unknown. We therefore analyzed a collection of cases for genomic aberrations using SNP arrays and targeted next-generation sequencing (tNGS) of oncogenes and tumor-suppressor genes. Recurrent gains of chromosome 1q and a small region of chromosome 4 encompassing KDR and KIT were identified by SNP array analysis. Repeatedly mutated genes identified by tNGS were KDR with different nonsynonymous mutations, MLL2 with different nonsense mutations, and PLCG1 with a recurrent nonsynonymous mutation (R707Q) in the highly conserved autoinhibitory SH2 domain in three of 10 cases. PLCγ1 is usually activated by Y783 phosphorylation and activates protein kinase C and Ca(2+)-dependent second messengers, with effects on cellular proliferation, migration, and invasiveness. Ectopic expression of the PLCγ1-R707Q mutant in endothelial cells revealed reduced PLCγ1-Y783 phosphorylation with concomitant increased c-RAF/MEK/ERK1/2 phosphorylation, increased IP3 amounts, and increased Ca(2+)-dependent calcineurin activation compared with ectopic expressed PLCγ1-wild-type. Furthermore, cofilin, whose activation is associated with actin skeleton reorganization, showed decreased phosphorylation, and thus activation after expression of PLCγ1-R707Q compared with PLCγ1-wild-type. At the cellular level, expression of PLCγ1-R707Q in endothelial cells had no influence on proliferation rate, but increased apoptosis resistance and migration and invasiveness in in vitro assays. Together, these findings indicate that the PLCγ1-R707Q mutation causes constitutive activation of PLCγ1 and may represent an alternative way of activation of KDR/PLCγ1 signaling besides KDR activation in angiosarcomas, with implications for VEGF/KDR targeted therapies.
Collapse
Affiliation(s)
- Kristin Kunze
- Department of Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Tilmann Spieker
- Institute for Pathology, St. Francis Hospital, Münster, Germany
| | - Ulrike Gamerdinger
- Department of Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Kerstin Nau
- Department of Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Johannes Berger
- Department of Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Thomas Dreyer
- Department of Pathology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Jürgen R Sindermann
- Department of Cardiothoracic Surgery, Division of Cardiac Surgery, University Hospital Münster, Münster, Germany
| | - Andreas Hoffmeier
- Department of Cardiothoracic Surgery, Division of Cardiac Surgery, University Hospital Münster, Münster, Germany
| | | | - Andreas Bräuninger
- Department of Pathology, Justus-Liebig-University Giessen, Giessen, Germany.
| |
Collapse
|
27
|
Csató V, Pető A, Koller Á, Édes I, Tóth A, Papp Z. Hydrogen peroxide elicits constriction of skeletal muscle arterioles by activating the arachidonic acid pathway. PLoS One 2014; 9:e103858. [PMID: 25093847 PMCID: PMC4122381 DOI: 10.1371/journal.pone.0103858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 07/02/2014] [Indexed: 02/07/2023] Open
Abstract
Aims The molecular mechanisms of the vasoconstrictor responses evoked by hydrogen peroxide (H2O2) have not been clearly elucidated in skeletal muscle arterioles. Methods and Results Changes in diameter of isolated, cannulated and pressurized gracilis muscle arterioles (GAs) of Wistar-Kyoto rats were determined under various test conditions. H2O2 (10–100 µM) evoked concentration-dependent constrictions in the GAs, which were inhibited by endothelium removal, or by antagonists of phospholipase A (PLA; 100 µM 7,7-dimethyl-(5Z,8Z)-eicosadienoic acid), protein kinase C (PKC; 10 µM chelerythrine), phospholipase C (PLC; 10 µM U-73122), or Src family tyrosine kinase (Src kinase; 1 µM Src Inhibitor-1). Antagonists of thromboxane A2 (TXA2; 1 µM SQ-29548) or the non-specific cyclooxygenase (COX) inhibitor indomethacin (10 µM) converted constrictions to dilations. The COX-1 inhibitor (SC-560, 1 µM) demonstrated a greater reduction in constriction and conversion to dilation than that of COX-2 (celecoxib, 3 µM). H2O2 did not elicit significant changes in arteriolar Ca2+ levels measured with Fura-2. Conclusions These data suggest that H2O2 activates the endothelial Src kinase/PLC/PKC/PLA pathway, ultimately leading to the synthesis and release of TXA2 by COX-1, thereby increasing the Ca2+ sensitivity of the vascular smooth muscle cells and eliciting constriction in rat skeletal muscle arterioles.
Collapse
Affiliation(s)
- Viktória Csató
- Division of Clinical Physiology, Institute of Cardiology, University of Debrecen, Debrecen, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Pető
- Division of Clinical Physiology, Institute of Cardiology, University of Debrecen, Debrecen, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Ákos Koller
- Department of Pathophysiology and Gerontology, University of Pécs, Pécs, Hungary
- Department of Pathophysiology, Semmelweis University, Budapest, Hungary
- Department of Physiology, New York Medical College, Valhalla, New York, United States of America
| | - István Édes
- Division of Clinical Physiology, Institute of Cardiology, University of Debrecen, Debrecen, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Tóth
- Division of Clinical Physiology, Institute of Cardiology, University of Debrecen, Debrecen, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Institute of Cardiology, University of Debrecen, Debrecen, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, Hungary
- * E-mail:
| |
Collapse
|
28
|
Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal 2014; 20:1126-67. [PMID: 23991888 PMCID: PMC3929010 DOI: 10.1089/ars.2012.5149] [Citation(s) in RCA: 3119] [Impact Index Per Article: 283.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Abstract Reactive oxygen species (ROS) are key signaling molecules that play an important role in the progression of inflammatory disorders. An enhanced ROS generation by polymorphonuclear neutrophils (PMNs) at the site of inflammation causes endothelial dysfunction and tissue injury. The vascular endothelium plays an important role in passage of macromolecules and inflammatory cells from the blood to tissue. Under the inflammatory conditions, oxidative stress produced by PMNs leads to the opening of inter-endothelial junctions and promotes the migration of inflammatory cells across the endothelial barrier. The migrated inflammatory cells not only help in the clearance of pathogens and foreign particles but also lead to tissue injury. The current review compiles the past and current research in the area of inflammation with particular emphasis on oxidative stress-mediated signaling mechanisms that are involved in inflammation and tissue injury.
Collapse
Affiliation(s)
- Manish Mittal
- 1 Department of Pharmacology, Center for Lung and Vascular Biology, University of Illinois College of Medicine, Chicago, Illinois
| | | | | | | | | |
Collapse
|
29
|
Collaboration of AMPK and PKC to induce phosphorylation of Ser413 on PIP5K1B resulting in decreased kinase activity and reduced PtdIns(4,5)P2 synthesis in response to oxidative stress and energy restriction. Biochem J 2014; 455:347-58. [PMID: 23909401 DOI: 10.1042/bj20130259] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The spatial and temporal regulation of the second messenger PtdIns(4,5)P2 has been shown to be crucial for regulating numerous processes in the cytoplasm and in the nucleus. Three isoforms of PIP5K1 (phosphatidylinositol 4-phosphate 5-kinase), A, B and C, are responsible for the regulation of the major pools of cellular PtdIns(4,5)P2. PIP5K1B is negatively regulated in response to oxidative stress although it remains unclear which pathways regulate its activity. In the present study, we have investigated the regulation of PIP5K1B by protein phosphorylation. Using MS analysis, we identified 12 phosphorylation sites on PIP5K1B. We developed a phospho-specific antibody against Ser413 and showed that its phosphorylation was increased in response to treatment of cells with phorbol ester, H2O2 or energy restriction. Using inhibitors, we define a stress-dependent pathway that requires the activity of the cellular energy sensor AMPK (AMP-activated protein kinase) and PKC (protein kinase C) to regulate Ser413 phosphorylation. Furthermore, we demonstrate that PKC can directly phosphorylate Ser413 in vitro. Mutation of Ser413 to aspartate to mimic serine phosphorylation decreased both PIP5K1B activity in vitro and PtdIns(4,5)P2 synthesis in vivo. Our studies show that collaboration between AMPK and PKC dictates the extent of Ser413 phosphorylation on PIP5K1B and regulates PtdIns(4,5)P2 synthesis.
Collapse
|
30
|
Aggarwal S, Gross CM, Sharma S, Fineman JR, Black SM. Reactive oxygen species in pulmonary vascular remodeling. Compr Physiol 2013; 3:1011-34. [PMID: 23897679 DOI: 10.1002/cphy.c120024] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The pathogenesis of pulmonary hypertension is a complex multifactorial process that involves the remodeling of pulmonary arteries. This remodeling process encompasses concentric medial thickening of small arterioles, neomuscularization of previously nonmuscular capillary-like vessels, and structural wall changes in larger pulmonary arteries. The pulmonary arterial muscularization is characterized by vascular smooth muscle cell hyperplasia and hypertrophy. In addition, in uncontrolled pulmonary hypertension, the clonal expansion of apoptosis-resistant endothelial cells leads to the formation of plexiform lesions. Based upon a large number of studies in animal models, the three major stimuli that drive the vascular remodeling process are inflammation, shear stress, and hypoxia. Although, the precise mechanisms by which these stimuli impair pulmonary vascular function and structure are unknown, reactive oxygen species (ROS)-mediated oxidative damage appears to play an important role. ROS are highly reactive due to their unpaired valence shell electron. Oxidative damage occurs when the production of ROS exceeds the quenching capacity of the antioxidant mechanisms of the cell. ROS can be produced from complexes in the cell membrane (nicotinamide adenine dinucleotide phosphate-oxidase), cellular organelles (peroxisomes and mitochondria), and in the cytoplasm (xanthine oxidase). Furthermore, low levels of tetrahydrobiopterin (BH4) and L-arginine the rate limiting cofactor and substrate for endothelial nitric oxide synthase (eNOS), can cause the uncoupling of eNOS, resulting in decreased NO production and increased ROS production. This review will focus on the ROS generation systems, scavenger antioxidants, and oxidative stress associated alterations in vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | | | |
Collapse
|
31
|
Domijan AM, Kovac S, Abramov AY. Lipid peroxidation is essential for phospholipase C activity and the inositol-trisphosphate-related Ca²⁺ signal. J Cell Sci 2013; 127:21-6. [PMID: 24198393 DOI: 10.1242/jcs.138370] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Reactive oxygen species (ROS) are produced in enzymatic and non-enzymatic reactions and have important roles in cell signalling but also detrimental effects. ROS-induced damage has been implicated in a number of neurological diseases; however, antioxidant therapies targeting brain diseases have been unsuccessful. Such failure might be related to inhibition of ROS-induced signalling in the brain. Using direct kinetic measures of lipid peroxidation in astrocytes and measurements of lipid peroxidation products in brain tissue, we here show that phospholipase C (PLC) preferentially cleaves oxidised lipids. Because of this, an increase in the rate of lipid peroxidation leads to increased Ca(2+) release from endoplasmic reticulum (ER) stores in response to physiological activation of purinoreceptors with ATP. Both vitamin E and its water-soluble analogue Trolox, potent ROS scavengers, were able to suppress PLC activity, therefore dampening intracellular Ca(2+) signalling. This implies that antioxidants can compromise intracellular Ca(2+) signalling through inhibition of PLC, and that PLC plays a dual role - signalling and antioxidant defence.
Collapse
|
32
|
Frindt G, Li H, Sackin H, Palmer LG. Inhibition of ROMK channels by low extracellular K+ and oxidative stress. Am J Physiol Renal Physiol 2013; 305:F208-15. [PMID: 23678039 DOI: 10.1152/ajprenal.00185.2013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We tested the hypothesis that low luminal K⁺ inhibits the activity of ROMK channels in the rat cortical collecting duct. Whole-cell voltage-clamp measurements of the component of outward K⁺ current inhibited by the bee toxin Tertiapin-Q (ISK) showed that reducing the bath concentration ([K⁺]o) to 1 mM resulted in a decline of current over 2 min compared with that observed at 10 mM [K⁺]o. However, maintaining tubules in 1 mM [K⁺]o without establishing whole-cell clamp conditions did not affect ISK. The [K⁺]o-dependent decline was not prevented by increasing cytoplasmic-side pH or by inhibition of phosphatase activity. It was, however, abolished by the inclusion of 0.5 mM DTT in the pipette solution to prevent oxidation of the intracellular environment. Conversely, treatment of intact tubules with the oxidant H₂O₂ (100 μM) decreased ISK in a [K⁺]o-dependent manner. Treatment of the tubules with the phospholipase C inhibitor U73122 prevented the effect of low [K⁺]o, suggesting the involvement of this enzyme in the process. We examined these effects further using Xenopus oocytes expressing ROMK2 channels. A 50-min exposure to the permeant oxidizing agent tert-butyl hydroperoxide (t-BHP; 500 μM) did not affect outward K⁺ currents with [K⁺]o = 10 mM but reduced currents by 50% with [K⁺]o = 1 mM and by 75% with [K⁺]o = 0.1 mM. Pretreatment of the oocytes with U73122 prevented the effects of t-BHP. Under conditions of low dietary K intake, K⁺ secretion by distal nephron segments may be suppressed by a combination of low luminal [K⁺]o and oxidative stress.
Collapse
Affiliation(s)
- Gustavo Frindt
- Department of Physiology and Biophysics, Weill-Cornell Medical College, New York, NY 10065, USA
| | | | | | | |
Collapse
|
33
|
Matsumoto K, Shimizu N. Activation of the phospholipase C signaling pathway in nerve growth factor-treated neurons by carbon nanotubes. Biomaterials 2013; 34:5988-94. [PMID: 23669261 DOI: 10.1016/j.biomaterials.2013.04.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 04/21/2013] [Indexed: 01/30/2023]
Abstract
Low concentrations of carbon nanotubes (CNTs) promoted the number of nerve growth factor (NGF)-treated neurons with neurite outgrowth by activating extracellular signal-regulated kinase (ERK), even when MEK inhibitor was added to the neuron culture medium. We speculated that CNTs may activate ERK through the phospholipase C (PLC) signaling pathway independent of the Ras/Raf/MEK cascade involved in the ERK signaling pathway. CNTs enhanced phosphorylation of PLC-γ1 in NGF-treated neurons but failed to increase the number and length of neurites of NGF-treated neurons with neurite outgrowth when a PLC inhibitor, an inositol triphosphate receptor (IP3R) inhibitor, or an inhibitor of protein kinase C (PKC) in the PLC signaling pathway were added to the neuron culture medium. Furthermore, intracellular Ca(++) levels of cells treated with CNTs+NGF were higher than those of cells treated with NGF alone. Although the combination of CNTs and NGF increased the concentration of phosphorylated ERK (p-ERK) in MEK inhibitor-treated neurons, CNTs did not induce phosphorylation of ERK in PLC inhibitor-treated neurons. These data suggest that PKC in the PLC signaling pathway may activate ERK independent of the Ras/Raf/MEK cascade. In summary, we identified a role of PLC signaling in mediating neurite outgrowth of NGF-treated neurons in the presence of CNTs.
Collapse
Affiliation(s)
- Kotaro Matsumoto
- Bio-Nano Electronics Research Center, Toyo University, 2100 Kujirai, Kawagoe-shi, Saitama 350-8585, Japan
| | | |
Collapse
|
34
|
Drigotas M, Affolter A, Mann WJ, Brieger J. Reactive oxygen species activation of MAPK pathway results in VEGF upregulation as an undesired irradiation response. J Oral Pathol Med 2013; 42:612-9. [PMID: 23480207 DOI: 10.1111/jop.12056] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND Radioresistance limits the effectiveness of radiotherapy in head and neck squamous cell carcinoma. We previously demonstrated post-radiogenic mitogen-activated protein kinase (MAPK) pathway activation and vascular endothelial growth factor (VEGF) release resulting in reduced tumor cell response. Here, we examined the association of this mechanism with the induction of reactive oxygen species (ROS) under irradiation (IR). METHODS Intracellular ROS after IR were measured. We modeled radiation-induced ROS by exposure of two SCC lines to H2 O2 and evaluated the impact of irradiation and ROS on ERK phosphorylation by Western blot, immunohistochemistry, and ELISA. RESULTS We found elevated pERK levels after treatment with IR and H2 O2 , which could be distinctly suppressed by U0126. Immunohistochemistry and ELISA revealed increased intracellular VEGF levels after H2 O2 application. CONCLUSIONS Our data show that irradiation-induced ROS activate the MAPK pathway and release of VEGF. As VEGF is known to be released after cellular distress resulting in cytoprotection, the described mechanism is potentially of importance for therapy success.
Collapse
Affiliation(s)
- Martynas Drigotas
- Department of Otorhinolaryngology, Head and Neck Surgery, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | | | | |
Collapse
|
35
|
Yadav VR, Song T, Joseph L, Mei L, Zheng YM, Wang YX. Important role of PLC-γ1 in hypoxic increase in intracellular calcium in pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2012. [PMID: 23204067 DOI: 10.1152/ajplung.00310.2012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An increase in intracellular calcium concentration ([Ca(2+)](i)) in pulmonary arterial smooth muscle cells (PASMCs) induces hypoxic cellular responses in the lungs; however, the underlying molecular mechanisms remain incompletely understood. We report, for the first time, that acute hypoxia significantly enhances phospholipase C (PLC) activity in mouse resistance pulmonary arteries (PAs), but not in mesenteric arteries. Western blot analysis and immunofluorescence staining reveal the expression of PLC-γ1 protein in PAs and PASMCs, respectively. The activity of PLC-γ1 is also augmented in PASMCs following hypoxia. Lentiviral shRNA-mediated gene knockdown of mitochondrial complex III Rieske iron-sulfur protein (RISP) to inhibit reactive oxygen species (ROS) production prevents hypoxia from increasing PLC-γ1 activity in PASMCs. Myxothiazol, a mitochondrial complex III inhibitor, reduces the hypoxic response as well. The PLC inhibitor U73122, but not its inactive analog U73433, attenuates the hypoxic vasoconstriction in PAs and hypoxic increase in [Ca(2+)](i) in PASMCs. PLC-γ1 knockdown suppresses its protein expression and the hypoxic increase in [Ca(2+)](i). Hypoxia remarkably increases inositol 1,4,5-trisphosphate (IP(3)) production, which is blocked by U73122. The IP(3) receptor (IP(3)R) antagonist 2-aminoethoxydiphenyl borate (2-APB) or xestospongin-C inhibits the hypoxic increase in [Ca(2+)](i). PLC-γ1 knockdown or U73122 reduces H(2)O(2)-induced increase in [Ca(2+)](i) in PASMCs and contraction in PAs. 2-APB and xestospongin-C produce similar inhibitory effects. In conclusion, our findings provide novel evidence that hypoxia activates PLC-γ1 by increasing RISP-dependent mitochondrial ROS production in the complex III, which causes IP(3) production, IP(3)R opening, and Ca(2+) release, playing an important role in hypoxic Ca(2+) and contractile responses in PASMCs.
Collapse
Affiliation(s)
- Vishal R Yadav
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Yang YR, Choi JH, Chang JS, Kwon HM, Jang HJ, Ryu SH, Suh PG. Diverse cellular and physiological roles of phospholipase C-γ1. Adv Biol Regul 2012; 52:138-151. [PMID: 21964416 DOI: 10.1016/j.advenzreg.2011.09.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 09/19/2011] [Indexed: 05/31/2023]
Affiliation(s)
- Yong Ryoul Yang
- School of Nano-Biotechnology and Chemical Engineering, Ulsan National Institute of Science and Technology, Ulsan 689-798, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
37
|
PLCγ is required for RhoGDI2-mediated cisplatin resistance in gastric cancer. Biochem Biophys Res Commun 2011; 414:575-80. [PMID: 21986528 DOI: 10.1016/j.bbrc.2011.09.121] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 09/22/2011] [Indexed: 01/23/2023]
Abstract
Rho GDP dissociation inhibitor 2 (RhoGDI2) is a regulator of the Rho family GTPases. Recent work from our laboratory suggests that RhoGDI2 expression potentially enhances resistance to cisplatin as well as promotes tumor growth and malignant progression in gastric cancer. In this study, we demonstrate that phospholipase C-gamma (PLCγ) is required for RhoGDI2-mediated cisplatin resistance and cancer cell invasion in gastric cancer. The levels of phosphorylated PLCγ are markedly enhanced in RhoGDI2-overexpressing SNU-484 cells and, by contrast, repressed in RhoGDI2-depleted MKN-28 cells. Depletion of PLCγ expression or inhibition of its activity not only significantly increases cisplatin-induced apoptosis but also suppresses the invasive ability of RhoGDI2-overexpressing SNU-484 cells. Taken together, our results suggest that PLCγ plays a key role in RhoGDI2-mediated cisplatin resistance and cell invasion in gastric cancer cells.
Collapse
|
38
|
Runchel C, Matsuzawa A, Ichijo H. Mitogen-activated protein kinases in mammalian oxidative stress responses. Antioxid Redox Signal 2011; 15:205-18. [PMID: 21050144 DOI: 10.1089/ars.2010.3733] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
All aerobic organisms are exposed to oxidative stress during their lifetime and are required to respond appropriately for maintenance of their survival and homeostasis. Sustained exposure to oxidative stress has devastating effects in organisms, and, not surprisingly, oxidative stress has been implicated in numerous human diseases. Therefore, an understanding of how mammals respond to oxidative stress is crucial both biologically and clinically. Intracellular signaling pathways, which are activated in response to excessive oxygen radicals, play essential roles in overcoming oxidative stress. The mitogen-activated protein kinase (MAPK) signaling pathways are involved in diverse physiological processes, and are critical for induction of oxidative stress responses. In this review, we will discuss the physiological roles of MAPKs in oxidative stress, the upstream signaling pathways leading to MAPK activation, their regulation, and the MAPK downstream substrates, with a focus on mammalian systems.
Collapse
Affiliation(s)
- Christopher Runchel
- Laboratory of Cell Signaling, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Japan
| | | | | |
Collapse
|
39
|
Hemi R, Yochananov Y, Barhod E, Kasher-Meron M, Karasik A, Tirosh A, Kanety H. p38 mitogen-activated protein kinase-dependent transactivation of ErbB receptor family: a novel common mechanism for stress-induced IRS-1 serine phosphorylation and insulin resistance. Diabetes 2011; 60:1134-45. [PMID: 21386087 PMCID: PMC3064087 DOI: 10.2337/db09-1323] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Stress stimuli such as tumor necrosis factor (TNF) have been shown to induce insulin receptor substrate (IRS)-1 serine phosphorylation and insulin resistance by transactivation of ErbB receptors. We aimed at elucidating the potential role of p38 mitogen-activated protein kinase (p38MAPK) in mediating stress-induced ErbB receptors activation. RESEARCH DESIGN AND METHODS p38MAPK effect on ErbBs transactivation and insulin signaling was assessed in Fao or HepG2 cells, exposed to stress stimuli, and on metabolic parameters in ob/ob and C57/BL6 mice. RESULTS High-fat diet-fed mice and ob/ob mice exhibited elevated hepatic p38MAPK activation associated with glucose intolerance and hyperinsulinemia. Liver expression of dominant-negative (DN)-p38MAPKα in ob/ob mice reduced fasting insulin levels and improved glucose tolerance, whereas C57/BL6 mice overexpressing wild-type p38MAPKα exhibited enhanced IRS-1 serine phosphorylation and reduced insulin-stimulated IRS-1 tyrosine phosphorylation. Fao or HepG2 cells exposed to TNF, anisomycin, or sphingomyelinase demonstrated rapid transactivation of ErbB receptors leading to PI3-kinase/Akt activation and IRS-1 serine phosphorylation. p38MAPK inhibition either by SB203580, by small interfering RNA, or by DN-p38MAPKα decreased ErbB receptors transactivation and IRS-1 serine phosphorylation and partially restored insulin-stimulated IRS-1 tyrosine phosphorylation. When cells were incubated with specific ErbB receptors antagonists or in cells lacking ErbB receptors, anisomycin- and TNF-induced IRS-1 serine phosphorylation was attenuated, despite intact p38MAPK activation. The stress-induced p38MAPK activation leading to ErbB receptors transactivation was associated with intracellular reactive oxygen species generation and was attenuated by treatment with antioxidants. CONCLUSIONS Hepatic p38MAPK is activated following various stress stimuli. This event is upstream to ErbB receptors transactivation and plays an important role in stress-induced IRS-1 serine phosphorylation and insulin resistance.
Collapse
Affiliation(s)
- Rina Hemi
- Institute of Endocrinology, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Yafit Yochananov
- Institute of Endocrinology, Chaim Sheba Medical Center, Tel-Hashomer, Israel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Ehud Barhod
- Institute of Endocrinology, Chaim Sheba Medical Center, Tel-Hashomer, Israel
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Michal Kasher-Meron
- Institute of Endocrinology, Chaim Sheba Medical Center, Tel-Hashomer, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avraham Karasik
- Institute of Endocrinology, Chaim Sheba Medical Center, Tel-Hashomer, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amir Tirosh
- Institute of Endocrinology, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Hannah Kanety
- Institute of Endocrinology, Chaim Sheba Medical Center, Tel-Hashomer, Israel
- The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Corresponding author: Hannah Kanety,
| |
Collapse
|
40
|
Heo J. Redox control of GTPases: from molecular mechanisms to functional significance in health and disease. Antioxid Redox Signal 2011; 14:689-724. [PMID: 20649471 DOI: 10.1089/ars.2009.2984] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Small GTPases, including the proto-oncoprotein Ras and Rho GTPases, are involved in various cellular signaling events. Some of these small GTPases are redox sensitive, including Ras, Rho, Ran, Dexras1, and Rhes GTPases. Thus, the redox-mediated regulation of these GTPases often determines the course of their cellular signaling cascades. This article takes into consideration the application of Marcus theory to potential redox-based molecular mechanisms in the regulation of these redox-sensitive GTPases and the relevance of such mechanisms to a specific redox-sensitive motif. The discussion also takes into account various diseases, including cancers, heart, and neuronal disorders, that are often linked with the dysregulation of the redox signaling cascades associated with these redox-sensitive GTPases.
Collapse
Affiliation(s)
- Jongyun Heo
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, Texas 76019, USA.
| |
Collapse
|
41
|
Wei L, Lu N, Dai Q, Rong J, Chen Y, Li Z, You Q, Guo Q. Different apoptotic effects of wogonin via induction of H2O2 generation and Ca2+ overload in malignant hepatoma and normal hepatic cells. J Cell Biochem 2010; 111:1629-41. [DOI: 10.1002/jcb.22898] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
42
|
The adaptor protein SH2B1β reduces hydrogen peroxide-induced cell death in PC12 cells and hippocampal neurons. J Mol Signal 2010; 5:17. [PMID: 20868529 PMCID: PMC2954984 DOI: 10.1186/1750-2187-5-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 09/27/2010] [Indexed: 11/25/2022] Open
Abstract
Background SH2B1β is a signaling adaptor protein that has been shown to promote neuronal differentiation in PC12 cells and is necessary for the survival of sympathetic neurons. However, the mechanism by which SH2B1β may influence cell survival is not known. Results In this study, we investigated the role of SH2B1β in oxidative stress-induced cell death. Our results suggest that overexpressing SH2B1β reduced H2O2-induced, caspase 3-dependent apoptosis in PC12 cells and hippocampal neurons. In response to H2O2, overexpressing SH2B1β enhanced PI3K (phosphatidylinositol 3-kinas)-AKT (protein kinase B) and MEK (MAPK/ERK kinase)-extracellular-signal regulated kinases 1 and 2 (ERK1/2) signaling pathways. We further demonstrated that SH2B1β was able to reduce H2O2-induced nuclear localization of FoxO1 and 3a transcription factors, which lie downstream of PI3K-AKT and MEK-ERK1/2 pathways. Moreover, overexpressing SH2B1β reduced the expression of Fas ligand (FasL), one of the target genes of FoxOs. Conclusions Overexpressing the adaptor protein SH2B1β enhanced H2O2-induced PI3K-AKT and MEK-ERK1/2 signaling, reduced nucleus-localized FoxOs and the expression of a pro-apoptotic gene, FasL.
Collapse
|
43
|
Thubagere A, Reinhard BM. Nanoparticle-induced apoptosis propagates through hydrogen-peroxide-mediated bystander killing: insights from a human intestinal epithelium in vitro model. ACS NANO 2010; 4:3611-22. [PMID: 20560658 DOI: 10.1021/nn100389a] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The ability to assess the risks of human exposure to engineered nanomaterials requires fundamental understanding of the fate and potential cytotoxicity of nonbiodegradable nanoparticles, for instance, after oral uptake. In this study, we quantify the impact of nanoparticles with low chemical toxicity on the intestinal membrane in a human intestinal in vitro model. Differentiated human colorectal adenocarcinoma cells, Caco-2, were cultured on a permeable support where they form an epithelial monolayer separating an apical and basal compartment. This model system allows a systematic characterization of the effect of nanoparticles on the cell viability as a function of size, surface chemistry, concentration, and incubation time. We used polystyrene (PS) nanoparticles (20 and 40 nm diameter) with two different surface chemistries (carboxylic acid and amines). The experiments performed show a strong decrease in cell viability as a response to nanoparticle exposure. Incubation times of <or=4 h are sufficient to induce dramatic losses in cell viability after an additional induction period of 4-12 h. Mapping the temporospatial distribution of dead cells in the Caco-2 cell monolayer using optical microscopy reveals that the nanoparticles induce apoptosis in individual cells, which then propagate across the cell monolayer through a "bystander killing effect". Addition of catalase, which selectively decomposes hydrogen peroxide, leads to a significant decrease in apoptosis levels, indicating that hydrogen peroxide causes the spread of apoptosis across the monolayer. Our findings confirm that ingested nonbiodegradable nanoparticles represent a potential health risk due to their detrimental impact on the intestinal membrane by destroying their barrier protection capability over time.
Collapse
Affiliation(s)
- Anupama Thubagere
- Department of Chemistry and The Photonics Center, Boston University, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
44
|
Cavet ME, Smolock EM, Menon P, Konishi A, Korshunov VA, Berk BC. Gas6-Axl pathway: the role of redox-dependent association of Axl with nonmuscle myosin IIB. Hypertension 2010; 56:105-11. [PMID: 20479336 DOI: 10.1161/hypertensionaha.109.144642] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In vascular smooth muscle cells, Axl is a key receptor tyrosine kinase, because it is upregulated in injury, increases migration and neointima formation, and is activated by reactive oxygen species. Reaction of glutathione with cysteine residues (termed "glutathiolation") is an important posttranslational redox modification that may alter protein activity and protein-protein interactions. To investigate the mechanisms by which reactive oxygen species increase Axl-dependent vascular smooth muscle cell function we assayed for glutathiolated proteins that associated with Axl in a redox-dependent manner. We identified glutathiolated nonmuscle myosin heavy chain (MHC)-IIB as a novel Axl interacting protein. This interaction was specific in that other myosins did not interact with Axl. The endogenous ligand for Axl, Gas6, increased production of reactive oxygen species in vascular smooth muscle cells and also increased the association of Axl with MHC-IIB. Antioxidants ebselen and N-acetylcysteine decreased the association of Axl with MHC-IIB in response to both Gas6 and reactive oxygen species. Blocking the Axl-MHC-IIB interaction with the specific myosin II inhibitor blebbistatin decreased phosphorylation of Axl and activation of extracellular signal-regulated kinase 1/2 and Akt. Association of MHC-IIB with Axl was increased in balloon-injured rat carotid vessels. Finally, expression of MHC-IIB was upregulated in the neointima of the carotid artery after balloon injury similar to upregulation of Axl protein expression, as shown in our previous studies. These results demonstrate a novel interaction between Axl and MHC-IIB in response to reactive oxygen species. This interaction provides a direct link between Axl and molecular motors crucial for directed cell migration, which may mediate increased migration in vascular dysfunction.
Collapse
Affiliation(s)
- Megan E Cavet
- Aab Cardiovascular Research Institute, University of Rochester, Box 679, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
45
|
Wang ZJ, Sun P, Xing W, Pan C, Lin DH, Wang WH. Decrease in dietary K intake stimulates the generation of superoxide anions in the kidney and inhibits K secretory channels in the CCD. Am J Physiol Renal Physiol 2010; 298:F1515-22. [PMID: 20357031 DOI: 10.1152/ajprenal.00502.2009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously demonstrated that K depletion inhibited ROMK-like small-conductance K channels (SK) in the cortical collecting duct (CCD) and that the effect was mediated by superoxide anions that stimulated Src family protein tyrosine kinase (PTK) and mitogen-activated protein kinase (MAPK) (51). However, because animals on a K-deficient diet had a severe hypokalemia, superoxide-dependent signaling may not regulate ROMK channels under physiological conditions with a normal plasma K concentration. In the present study, we used the patch-clamp technique and Western blot to examine the effect of a moderate K restriction on ROMK-like SK channels and the role of PTK and MAPK in regulating apical K channels in the CCD of animals on a low-K diet (LK; 0.1% K). Rats and mice fed a LK diet for 7 days had a normal plasma K concentration. However, a LK intake increased the expression of angiotensin II type 1 receptor in the kidney. Moreover, patch-clamp experiments demonstrated that LK intake decreased the probability finding SK channels and channel activity defined by NP(o) (a product of channel number and open probability) in the CCD of both rat and mouse kidneys. Also, LK intake significantly stimulated the production of superoxide anions in the renal cortex and outer medulla in both rats and mice and increased superoxide level in the rat CCD. Moreover, LK intake augments the phosphorylation of p38 and ERK MAPK, the expression of c-Src and tyrosine phosphorylation of ROMK channels. However, treatment of animals with tempol abolished the effect of LK intake on MAPK and c-Src and increased ROMK channel activity in comparing with those of nontreated rats on a LK diet. Inhibiting p38 and ERK with SB202190 and PD98059 significantly stimulated SK in the CCD in rats on a LK diet. In addition, inhibition of PTK with herbimycin A activated SK channels in the CCD from rats on a LK diet. We conclude that LK intake stimulates the generation of superoxide anion and related products and that MAPK and Src family PTK play a physiological role in inhibiting apical K channels in the principal cells in response to LK intake.
Collapse
Affiliation(s)
- Zhi-Jian Wang
- Department of Pharmacology, New York Medical College, Valhalla, New York 10595, USA
| | | | | | | | | | | |
Collapse
|
46
|
Okada M, Taguchi K, Maekawa S, Fukami K, Yagisawa H. Calcium fluxes cause nuclear shrinkage and the translocation of phospholipase C-delta1 into the nucleus. Neurosci Lett 2010; 472:188-93. [PMID: 20138965 DOI: 10.1016/j.neulet.2010.01.081] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 01/22/2010] [Accepted: 01/31/2010] [Indexed: 12/28/2022]
Abstract
Phospholipase C-delta1 (PLCdelta1) is the most fundamental form of the eukaryotic PLC and thought to play important roles in the regulation of cells. We previously reported that PLCdelta1 shuttles between the cytoplasm and nucleus, and an influx of Ca2+ triggers the nuclear import of PLCdelta1 via Ca2+-dependent interaction with importin beta1, although the physiological meaning of this is unclear. Here we have examined the distribution of PLCdelta1 using primary cultures of rat hippocampal neurons. Treatment of 7DIV neurons with ionomycin or thapsigargin caused the nuclear localization of PLCdelta1 as has been observed in other cell lines. Similar results were obtained with neurons treated with glutamate, suggesting that the nuclear localization of PLCdelta1 plays some roles in excitotoxicity associated with ischemic stress. Generally, cells undergoing ischemic or hypoxic cell death show nuclear shrinkage. We confirmed that a massive influx of Ca2+ caused similar results. Furthermore, overexpression of GFP-PLCdelta1 facilitated ionomycin-induced nuclear shrinkage in embryonic fibroblasts derived from PLCdelta1 gene-knockout mice (PLCdelta1KO-MEF). By contrast, an E341A mutant that cannot bind with importin beta1 and be imported into the nucleus by ionomycin and also lacks enzymatic activity did not cause nuclear shrinkage in PLCdelta1KO-MEF. Nuclear translocation and the PLC activity of PLCdelta1, therefore, may regulate the nuclear shape by controlling the nuclear scaffold during stress-induced cell death caused by high levels of Ca2+.
Collapse
Affiliation(s)
- Masashi Okada
- Graduate School of Life Science, University of Hyogo, 3-2-1 Kouto, Kamigori, Hyogo 678-1297, Japan
| | | | | | | | | |
Collapse
|
47
|
Hemalatha T, Tiwari M, Balachandran C, Manohar BM, Puvanakrishnan R. Platelet-derived endothelial cell growth factor mediates angiogenesis and antiapoptosis in rat aortic endothelial cells. Biochem Cell Biol 2009; 87:883-93. [DOI: 10.1139/o09-056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study explores the angiogenic and antiapoptotic activities of platelet-derived endothelial cell growth factor (PDECGF) in rat aortic endothelial cells. The effects of PDECGF on rat aortic endothelial cell (RAEC) proliferation, migration, chemotaxis, and tubule formation were investigated in vitro at various concentrations viz., 1, 2, 4, 8, 16, and 32 ng·mL–1 on endothelial cells. Endothelial cells were induced with hypoxic stress and the antiapoptotic effects of PDECGF were analysed by cell survival assay, fluorescence microscopy, cell viability assay, and flow cytometry. The results demonstrated the angiogenic potential of PDECGF on endothelial cells in a dose-dependent manner. PDECGF at 16 and 32 ng·mL–1 increased cell proliferation (>80%), induced cell migration (>4 fold), stimulated chemotaxis (>2 fold), and increased tubule formation (>3 fold) compared with the control. Studies on hypoxic stress revealed the antiapoptotic nature of PDECGF on endothelial cells. PDECGF treatment enhanced cell survival by 14%, as well as cell viability by 13%, and decreased the percentage of apoptotic cells by 13% as demonstrated by fluorescence-activated cell sorter studies (FACS). In conclusion, this study demonstrated the angiogenic and antiapoptotic potentials of PDECGF on RAEC.
Collapse
Affiliation(s)
- Thiagarajan Hemalatha
- Department of Biotechnology, Central Leather Research Institute, Adyar, Chennai, India
- Department of Veterinary Pathology, Madras Veterinary College, Vepery, Chennai, India
- Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University, Madhavaram Milk Colony, Chennai
| | - Mitali Tiwari
- Department of Biotechnology, Central Leather Research Institute, Adyar, Chennai, India
- Department of Veterinary Pathology, Madras Veterinary College, Vepery, Chennai, India
- Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University, Madhavaram Milk Colony, Chennai
| | - Chidambaram Balachandran
- Department of Biotechnology, Central Leather Research Institute, Adyar, Chennai, India
- Department of Veterinary Pathology, Madras Veterinary College, Vepery, Chennai, India
- Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University, Madhavaram Milk Colony, Chennai
| | - Bhakthavatsalam Murali Manohar
- Department of Biotechnology, Central Leather Research Institute, Adyar, Chennai, India
- Department of Veterinary Pathology, Madras Veterinary College, Vepery, Chennai, India
- Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University, Madhavaram Milk Colony, Chennai
| | - Rengarajulu Puvanakrishnan
- Department of Biotechnology, Central Leather Research Institute, Adyar, Chennai, India
- Department of Veterinary Pathology, Madras Veterinary College, Vepery, Chennai, India
- Centre for Animal Health Studies, Tamil Nadu Veterinary and Animal Sciences University, Madhavaram Milk Colony, Chennai
| |
Collapse
|
48
|
Klingelhöfer J, Møller HD, Sumer EU, Berg CH, Poulsen M, Kiryushko D, Soroka V, Ambartsumian N, Grigorian M, Lukanidin EM. Epidermal growth factor receptor ligands as new extracellular targets for the metastasis-promoting S100A4 protein. FEBS J 2009; 276:5936-48. [DOI: 10.1111/j.1742-4658.2009.07274.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
49
|
Sase H, Watabe T, Kawasaki K, Miyazono K, Miyazawa K. VEGFR2-PLCgamma1 axis is essential for endothelial specification of VEGFR2+ vascular progenitor cells. J Cell Sci 2009; 122:3303-11. [PMID: 19706681 DOI: 10.1242/jcs.049908] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) plays crucial roles in vasculogenesis, a process involving cell proliferation, migration and differentiation. However, the molecular mechanism by which VEGFR2 signaling directs vascular endothelial differentiation of VEGFR2(+) mesodermal progenitors is not well understood. In this study, we examined the signal transduction pathway downstream of VEGFR2 for endothelial differentiation using an in vitro differentiation system of mouse embryonic stem-cell-derived VEGFR2(+) cells. Using chimeric receptors composed of VEGFR2 and VEGFR3, the third member of the VEGFR family, we found that signaling through tyrosine 1175 (Y1175, corresponding to mouse Y1173) of VEGFR2 is crucial for two processes of endothelial differentiation: endothelial specification of VEGFR2(+) progenitors, and subsequent survival of endothelial cells (ECs). Furthermore, we found that phospholipase Cgamma1 (PLCgamma1), which interacts with VEGFR2 through phosphorylated Y1175, is an inducer of endothelial specification. In contrast to VEGFR2, VEGFR3 does not transmit a signal for endothelial differentiation of VEGFR2(+) cells. We found that VEGFR3 does not activate PLCgamma1, although VEGFR3 has the ability to support endothelial cell survival. Taken together, these findings indicate that VEGFR2-PLCgamma1 signal relay gives rise to the unique function of VEGFR2, thus enabling endothelial differentiation from vascular progenitors.
Collapse
Affiliation(s)
- Hitoshi Sase
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Hongo, Tokyo, Japan
| | | | | | | | | |
Collapse
|
50
|
Extracellular HspBP1 and Hsp72 synergistically activate epidermal growth factor receptor. Biol Cell 2009; 101:351-60. [PMID: 18986301 DOI: 10.1042/bc20080069] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND INFORMATION Heat-inducible Hsp72 is the founding member of the Hsp70 (heat shock proteins of 70 kDa) family of molecular chaperones. It is localized primarily in cytoplasm and nucleus but is also found extracellularly. The source of e-Hsp72 (extracellular Hsp72) is not precisely identified and may not be the same in every situation. A number of studies demonstrated that e-Hsp72 plays an important role in cell survival, tumour rejection and immune response. However, currently little is known about regulation of e-Hsp72 function. In cells, Hsp72 is controlled by co-chaperones. An abundant co-chaperone, HspBP1 (Hsp72-binding protein 1) was found extracellularly in the serum. In the present study we analysed the secretion and function of e-HspBP1 (extracellular HspBP1). RESULTS A431 human squamous carcinoma cells accumulated Hsp72 and HspBP1 in chromogranin A-positive granules following heat stress or in the presence of U73122, an inhibitor of phospholipase C. Following these treatments, A431 cells also increased the secretion of both proteins into the culture medium. The secreted e-Hsp72 and e-HspBP1 were co-immunoprecipitated from the conditioned medium. Purified recombinant HspBP1 augmented e-Hsp72-mediated phosphorylation of EGFR (epidermal growth factor receptor) and its down-stream targets, ERK1 (extracellular signal-regulated kinase 1) and ERK2 in a concentration-dependent manner. Finally, a HspBP1 N-terminal domain deletion mutant and boiled recombinant HspBP1 did not affect the e-Hsp72-mediated activity. CONCLUSIONS Heat stress and PLC (phospholipase C) inhibition result in the enhanced secretion of both Hsp72 and HspBP1. In an extracellular environment, the two chaperones interact both physically and functionally, leading to the activation of th EGFR-ERK1/2 signalling pathway. However, the magnitude of EGFR activation depends on the e-HspBP1/e-Hsp72 ratio in the medium. Extracellular chaperone-mediated activation of EGFR can provide a survival advantage to cells under heat shock and other stresses.
Collapse
|