151
|
Diaphragm Muscle Weakness Following Acute Sustained Hypoxic Stress in the Mouse Is Prevented by Pretreatment with N-Acetyl Cysteine. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4805493. [PMID: 29670681 PMCID: PMC5836441 DOI: 10.1155/2018/4805493] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/29/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022]
Abstract
Oxygen deficit (hypoxia) is a major feature of cardiorespiratory diseases characterized by diaphragm dysfunction, yet the putative role of hypoxic stress as a driver of diaphragm dysfunction is understudied. We explored the cellular and functional consequences of sustained hypoxic stress in a mouse model. Adult male mice were exposed to 8 hours of normoxia, or hypoxia (FiO2 = 0.10) with or without antioxidant pretreatment (N-acetyl cysteine, 200 mg/kg i.p.). Ventilation and metabolism were measured. Diaphragm muscle contractile function, myofibre size and distribution, gene expression, protein signalling cascades, and oxidative stress (TBARS) were determined. Hypoxia caused pronounced diaphragm muscle weakness, unrelated to increased respiratory muscle work. Hypoxia increased diaphragm HIF-1α protein content and activated MAPK, mTOR, Akt, and FoxO3a signalling pathways, largely favouring protein synthesis. Hypoxia increased diaphragm lipid peroxidation, indicative of oxidative stress. FoxO3 and MuRF-1 gene expression were increased. Diaphragm 20S proteasome activity and muscle fibre size and distribution were unaffected by acute hypoxia. Pretreatment with N-acetyl cysteine substantially enhanced cell survival signalling, prevented hypoxia-induced diaphragm oxidative stress, and prevented hypoxia-induced diaphragm dysfunction. Hypoxia is a potent driver of diaphragm weakness, causing myofibre dysfunction without attendant atrophy. N-acetyl cysteine protects the hypoxic diaphragm and may have application as a potential adjunctive therapy.
Collapse
|
152
|
Hare BD, Thornton TM, Rincon M, Golijanin B, King SB, Jaworski DM, Falls WA. Two Weeks of Variable Stress Increases Gamma-H2AX Levels in the Mouse Bed Nucleus of the Stria Terminalis. Neuroscience 2018; 373:137-144. [PMID: 29352998 DOI: 10.1016/j.neuroscience.2018.01.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/15/2017] [Accepted: 01/10/2018] [Indexed: 12/31/2022]
Abstract
Recent reports demonstrate that DNA damage is induced, and rapidly repaired, in circuits activated by experience. Moreover, stress hormones are known to slow DNA repair, suggesting that prolonged stress may result in persistent DNA damage. Prolonged stress is known to negatively impact physical and mental health; however, DNA damage as a factor in stress pathology has only begun to be explored. Histone H2A-X phosphorylated at serine 139 (γH2AX) is a marker of DNA double-strand breaks (DSB), a type of damage that may lead to cell death if unrepaired. We hypothesized that a 14-day period of variable stress exposure sufficient to alter anxiety-like behavior in male C57BL/6J mice would produce an increase in γH2AX levels in the bed nucleus of the stria terminalis (BNST), a region implicated in anxiety and stress regulation. We observed that 14 days of variable stress, but not a single stress exposure, was associated with increased levels of γH2AX 24 h after termination of the stress paradigm. Further investigation found that phosphorylation levels of a pair of kinases associated with the DNA damage response, glycogen synthase kinase 3 β (GSK3β) and p38 mitogen-activated protein kinase (MAPK) were also elevated following variable stress. Our results suggest that unrepaired DNA DSBs and/or repetitive attempted repair may represent an important component of the allostatic load that stress places on the brain.
Collapse
Affiliation(s)
- Brendan D Hare
- Department of Psychology, University of Vermont, Burlington, VT 05405, United States.
| | - Tina M Thornton
- Department of Medicine and Immunobiology, University of Vermont, College of Medicine, Burlington, VT 05405, United States
| | - Mercedes Rincon
- Department of Medicine and Immunobiology, University of Vermont, College of Medicine, Burlington, VT 05405, United States
| | - Borivoj Golijanin
- Department of Psychology, University of Vermont, Burlington, VT 05405, United States
| | - S Bradley King
- Department of Psychology, University of Vermont, Burlington, VT 05405, United States
| | - Diane M Jaworski
- Department of Neurological Sciences, University of Vermont, College of Medicine, Burlington, VT 05405, United States
| | - William A Falls
- Department of Psychology, University of Vermont, Burlington, VT 05405, United States
| |
Collapse
|
153
|
Souilhol C, Harmsen MC, Evans PC, Krenning G. Endothelial–mesenchymal transition in atherosclerosis. Cardiovasc Res 2018; 114:565-577. [DOI: 10.1093/cvr/cvx253] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Celine Souilhol
- Department of Infection, Immunity & Cardiovascular Disease (IICD), Faculty of Medicine, Dentistry & Health, Royal Hallamshire Hospital, University of Sheffield, Sheffield, UK
| | - Martin C Harmsen
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ Groningen, The Netherlands
| | - Paul C Evans
- Department of Infection, Immunity & Cardiovascular Disease (IICD), Faculty of Medicine, Dentistry & Health, Royal Hallamshire Hospital, University of Sheffield, Sheffield, UK
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ Groningen, The Netherlands
| |
Collapse
|
154
|
Failure to Inactivate Nuclear GSK3β by Ser 389-Phosphorylation Leads to Focal Neuronal Death and Prolonged Fear Response. Neuropsychopharmacology 2018; 43:393-405. [PMID: 28832021 PMCID: PMC5729567 DOI: 10.1038/npp.2017.187] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/25/2017] [Accepted: 08/17/2017] [Indexed: 12/19/2022]
Abstract
GSK3β plays an essential role in promoting cell death and is emerging as a potential target for neurological diseases. Understanding the mechanisms that control neuronal GSK3β is critical. A ubiquitous mechanism to repress GSK3β involves Akt-mediated phosphorylation of Ser9. Here we show that phosphorylation of GSK3β on Ser389 mediated by p38 MAPK specifically inactivates nuclear GSK3β in the cortex and hippocampus. Using GSK3β Ser389 to Ala mutant mice, we show that failure to inactivate nuclear GSK3β by Ser389 phosphorylation causes neuronal cell death in subregions of the hippocampus and cortex. Although this focal neuronal death does not impact anxiety/depression-like behavior or hippocampal-dependent spatial learning, it leads to an amplified and prolonged fear response. This phenotype is consistent with some aspects of post-traumatic stress disorder (PTSD). Our studies indicate that inactivation of nuclear GSK3β by Ser389 phosphorylation plays a key role in fear response, revealing new potential therapeutic approaches to target PTSD.
Collapse
|
155
|
Delayed neurochemical effects of prenatal exposure to MeHg in the cerebellum of developing rats. Toxicol Lett 2017; 284:161-169. [PMID: 29258870 DOI: 10.1016/j.toxlet.2017.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 12/03/2017] [Accepted: 12/09/2017] [Indexed: 01/09/2023]
Abstract
Human fetuses and neonates are particularly vulnerable to methylmercury (MeHg)-induced brain damage and are sensitive even to low exposure levels. Previous work of our group evidence that prenatal exposure to MeHg causes cognitive and behavioral alterations and disrupt hippocampus signaling. The current study aimed to investigate the effect of gestational exposure of rats to MeHg at low doses (1 or 2 mg/kg) on parameters of redox imbalance and key signaling pathways in the cerebellum of their offspring. Pregnant females received MeHg (treated group) or 0.9% saline water (control group) by gavage in alternated days from gestational day 5 (GD5) until parturition and analyzes were proceed in the cerebellum of 30-day-old pups. We found increased lipid peroxidation and protein carbonylation levels as well as decreased SH content in pups prenatally exposed to 2 mg/kg MeHg. In addition, misregulated SOD/catalase activities supported imbalanced redox equilibrium. We found decreased GSK3β(Ser9) phosphorylation, suggesting activation of this enzyme and dephosphorylation/inhibition of ERK1/2 and JNK pathways. Increased PKAα catalytic subunit could be upstream of hyperphosphorylated c-Raf(Ser259) and downregulated MAPK pathway. In addition, we found raised levels of the Ca2+-dependent protein phosphatase 2 B (PP2B). We also found preserved immunohistochemical staining for both glial fibrillary acidic protein (GFAP) and NeuN in MeHg-exposed pups. Western blot analysis showed unaltered levels of BAX/BCL-XL, BAD/BCL-2 and active caspase 3. Together, these findings support absence of reactive astrocytes, neuronal damage and apoptotic cell death in the cerebellum of MeHg treated pups. The present study provides evidence that prenatal exposure to MeHg leads to later redox imbalance and disrupted signaling mechanisms in the cerebellum of 30-day-old pups potentially predisposing them to long-lasting neurological impairments in CNS.
Collapse
|
156
|
Bhattacharya D, Majrashi M, Ramesh S, Govindarajulu M, Bloemer J, Fujihashi A, Crump BR, Hightower H, Bhattacharya S, Moore T, Suppiramaniam V, Dhanasekaran M. Assessment of the cerebellar neurotoxic effects of nicotine in prenatal alcohol exposure in rats. Life Sci 2017; 194:177-184. [PMID: 29225110 DOI: 10.1016/j.lfs.2017.12.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/27/2017] [Accepted: 12/06/2017] [Indexed: 01/12/2023]
Abstract
The adverse effects of prenatal nicotine and alcohol exposure on human reproductive outcomes are a major scientific and public health concern. In the United States, substantial percentage of women (20-25%) of childbearing age currently smoke cigarettes and consume alcohol, and only a small percentage of these individuals quit after learning of their pregnancy. However, there are very few scientific reports on the effect of nicotine in prenatal alcohol exposure on the cerebellum of the offspring. Therefore, this study was conducted to investigate the cerebellar neurotoxic effects of nicotine in a rodent model of Fetal Alcohol Spectrum Disorder (FASD). In this study, we evaluated the behavioral changes, biochemical markers of oxidative stress and apoptosis, mitochondrial functions and the molecular mechanisms associated with nicotine in prenatal alcohol exposure on the cerebellum. Prenatal nicotine and alcohol exposure induced oxidative stress, did not affect the mitochondrial functions, increased the monoamine oxidase activity, increased caspase expression and decreased ILK, PSD-95 and GLUR1 expression without affecting the GSK-3β. Thus, our current study of prenatal alcohol and nicotine exposure on cerebellar neurotoxicity may lead to new scientific perceptions and novel and suitable therapeutic actions in the future.
Collapse
Affiliation(s)
| | - Mohammed Majrashi
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Sindhu Ramesh
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Manoj Govindarajulu
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Jenna Bloemer
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Ayaka Fujihashi
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Bailee-Ryan Crump
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Harrison Hightower
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | | | - Timothy Moore
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | - Vishnu Suppiramaniam
- Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, USA
| | | |
Collapse
|
157
|
Zega K, Jovanovic VM, Vitic Z, Niedzielska M, Knaapi L, Jukic MM, Partanen J, Friedel RH, Lang R, Brodski C. Dusp16 Deficiency Causes Congenital Obstructive Hydrocephalus and Brain Overgrowth by Expansion of the Neural Progenitor Pool. Front Mol Neurosci 2017; 10:372. [PMID: 29170629 PMCID: PMC5684737 DOI: 10.3389/fnmol.2017.00372] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 10/26/2017] [Indexed: 12/22/2022] Open
Abstract
Hydrocephalus can occur in children alone or in combination with other neurodevelopmental disorders that are often associated with brain overgrowth. Despite the severity of these disorders, the molecular and cellular mechanisms underlying these pathologies and their comorbidity are poorly understood. Here, we studied the consequences of genetically inactivating in mice dual-specificity phosphatase 16 (Dusp16), which is known to negatively regulate mitogen-activated protein kinases (MAPKs) and which has never previously been implicated in brain development and disorders. Mouse mutants lacking a functional Dusp16 gene (Dusp16−/−) developed fully-penetrant congenital obstructive hydrocephalus together with brain overgrowth. The midbrain aqueduct in Dusp16−/− mutants was obstructed during mid-gestation by an expansion of neural progenitors, and during later gestational stages by neurons resulting in a blockage of cerebrospinal fluid (CSF) outflow. In contrast, the roof plate and ependymal cells developed normally. We identified a delayed cell cycle exit of neural progenitors in Dusp16−/− mutants as a cause of progenitor overproliferation during mid-gestation. At later gestational stages, this expanded neural progenitor pool generated an increased number of neurons associated with enlarged brain volume. Taken together, we found that Dusp16 plays a critical role in neurogenesis by balancing neural progenitor cell proliferation and neural differentiation. Moreover our results suggest that a lack of functional Dusp16 could play a central role in the molecular mechanisms linking brain overgrowth and hydrocephalus.
Collapse
Affiliation(s)
- Ksenija Zega
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Vukasin M Jovanovic
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Zagorka Vitic
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Magdalena Niedzielska
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Laura Knaapi
- Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Marin M Jukic
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Juha Partanen
- Department of Biosciences, University of Helsinki, Helsinki, Finland
| | - Roland H Friedel
- Departments of Neuroscience and Neurosurgery, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Claude Brodski
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
158
|
A partnership with the proteasome; the destructive nature of GSK3. Biochem Pharmacol 2017; 147:77-92. [PMID: 29102676 PMCID: PMC5954166 DOI: 10.1016/j.bcp.2017.10.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/31/2017] [Indexed: 12/19/2022]
Abstract
Glycogen Synthase Kinase-3 (GSK3) was originally reported as a key enzyme of glucose homeostasis through regulation of the rate of glycogen synthesis. It has subsequently been found to influence most cellular processes, including growth, differentiation and death, as part of its role in modulating response to hormonal, nutritional and cellular stress stimuli. More than 100 protein targets for GSK3 have been proposed although only a small fraction of these have been convincingly validated in physiological cell systems. The effects of GSK3 phosphorylation on substrates include alteration of enzyme activity, protein localisation, protein:protein interaction and protein stability. This latter form of regulation of GSK3 substrates is the focus of this review. There is an ever-growing list of GSK3 substrates that upon phosphorylation are targeted to the beta-transducin repeat containing protein (β-TrCP), thereby allowing ubiquitination of bound protein by cullin-1 and so initiating destruction at the proteasome. We propose the existence of a GSK3-β-TrCP ‘destruction hit-list’ that allows co-ordinated removal (or stabilisation) of a set of proteins with a common physiological purpose, through control of GSK3. We identify 29 proteins where there is relatively strong evidence for regulation by a GSK3-β-TrCP axis and note common features of regulation and pathophysiology. Furthermore, we assess the potential of pre-phosphorylation (priming) of these targets (normally a prerequisite for GSK3 recognition) to provide a second layer of regulation delineated by the priming kinase that allows GSK3 to mark them for destruction. Finally, we discuss whether this knowledge improves options for therapeutic intervention.
Collapse
|
159
|
Parveen S, Vedagiri D, Nair HG, Parthasarathy H, Harshan KH. Unconventional MAPK-GSK-3β Pathway Behind Atypical Epithelial-Mesenchymal Transition In Hepatocellular Carcinoma. Sci Rep 2017; 7:8842. [PMID: 28821798 PMCID: PMC5562823 DOI: 10.1038/s41598-017-09179-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/20/2017] [Indexed: 11/09/2022] Open
Abstract
We recently reported an atypical epithelial mesenchymal transition (EMT) in human hepatoma cell culture Huh7.5, which was non-responsive to the canonical EMT-transcription factors. Here we characterize major pathways regulating this atypical EMT through whole genome transcriptome profiling and molecular analysis, and identify a unique regulation of EMT by GSK-3β. Our analysis reveals remarkable suppression of several key liver-specific markers in Huh7.5M cells indicating that EMT not only changes the epithelial properties, but alters the characteristics associated with hepatocytes as well. One key finding of this study is that GSK-3β, a known antagonist to β-Catenin signaling and a major pro-apoptotic regulator, is critical for the maintenance of EMT in Huh7.5M cells as its inhibition reversed EMT. Importantly, through these studies we identify that maintenance of EMT by GSK-3β in Huh7.5M is regulated by p38MAPK and ERK1/2 that has not been reported elsewhere and is distinct from another metastatic non-hepatic cell line MDA-MB-231. These data showcase the existence of non-canonical mechanisms behind EMT. The atypicalness of this system underlines the existence of tremendous diversity in cancer-EMT and warrants the necessity to take a measured approach while dealing with metastasis and cancer drug resistance.
Collapse
Affiliation(s)
- Sana Parveen
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Dhiviya Vedagiri
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | - Hitha Gopalan Nair
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, 500007, India
| | | | | |
Collapse
|
160
|
In vivo regulation of glycogen synthase kinase 3β activity in neurons and brains. Sci Rep 2017; 7:8602. [PMID: 28819213 PMCID: PMC5561119 DOI: 10.1038/s41598-017-09239-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 07/19/2017] [Indexed: 12/31/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK3β) is a multifunctional protein kinase involved in many cellular activities including development, differentiation and diseases. GSK3β is thought to be constitutively activated by autophosphorylation at Tyr216 and inactivated by phosphorylation at Ser9. The GSK3β activity has previously been evaluated by inhibitory Ser9 phosphorylation, but it does not necessarily indicate the kinase activity itself. Here, we applied the Phos-tag SDS-PAGE technique to the analysis of GSK3β phosphoisotypes in cells and brains. There were three phosphoisotypes of GSK3β; double phosphorylation at Ser9 and Tyr216, single phosphorylation at Tyr216 and the nonphosphorylated isotype. Active GSK3β with phosphorylation at Tyr216 represented half or more of the total GSK3β in cultured cells. Although levels of phospho-Ser9 were increased by insulin treatment, Ser9 phosphorylation occurred only in a minor fraction of GSK3β. In mouse brains, GSK3β was principally in the active form with little Ser9 phosphorylation, and the phosphoisotypes of GSK3β changed depending on the regions of the brain, age, sex and disease conditions. These results indicate that the Phos-tag SDS-PAGE method provides a simple and appropriate measurement of active GSK3β in vivo, and the activity is regulated by the mechanism other than phosphorylation on Ser9.
Collapse
|
161
|
Arya R, Bassing CH. V(D)J Recombination Exploits DNA Damage Responses to Promote Immunity. Trends Genet 2017; 33:479-489. [PMID: 28532625 PMCID: PMC5499712 DOI: 10.1016/j.tig.2017.04.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 11/16/2022]
Abstract
It has been recognized for 40 years that the variable (diversity) joining [V(D)J] recombination-mediated assembly of diverse B and T lymphocyte antigen receptor (AgR) genes is not only essential for adaptive immunity, but also a risk for autoimmunity and lymphoid malignancies. Over the past few years, several studies have revealed that recombination-activating gene (RAG) endonuclease-induced DNA double-strand breaks (DSBs) transcend hazardous intermediates during antigen receptor gene assembly. RAG cleavage within the genomes of lymphocyte progenitors and immature lymphocytes regulates the expression of ubiquitous and lymphocyte-specific gene transcripts to control the differentiation and function of both adaptive and innate immune cell lineages. These unexpected discoveries raise important new questions that have broad implications for basic immunology research and the screening, diagnosis, and treatment of human immunological disease.
Collapse
Affiliation(s)
- Rahul Arya
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Craig H Bassing
- Division of Cancer Pathobiology, Department of Pathology and Laboratory Medicine, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
162
|
Molecular signaling cascades involved in nonmelanoma skin carcinogenesis. Biochem J 2017; 473:2973-94. [PMID: 27679857 DOI: 10.1042/bcj20160471] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/10/2016] [Indexed: 12/17/2022]
Abstract
Nonmelanoma skin cancer (NMSC) is the most common cancer worldwide and the incidence continues to rise, in part due to increasing numbers in high-risk groups such as organ transplant recipients and those taking photosensitizing medications. The most significant risk factor for NMSC is ultraviolet radiation (UVR) from sunlight, specifically UVB, which is the leading cause of DNA damage, photoaging, and malignant transformation in the skin. Activation of apoptosis following UVR exposure allows the elimination of irreversibly damaged cells that may harbor oncogenic mutations. However, UVR also activates signaling cascades that promote the survival of these potentially cancerous cells, resulting in tumor initiation. Thus, the UVR-induced stress response in the skin is multifaceted and requires coordinated activation of numerous pathways controlling DNA damage repair, inflammation, and kinase-mediated signal transduction that lead to either cell survival or cell death. This review focuses on the central signaling mechanisms that respond to UVR and the subsequent cellular changes. Given the prevalence of NMSC and the resulting health care burden, many of these pathways provide promising targets for continued study aimed at both chemoprevention and chemotherapy.
Collapse
|
163
|
Gowda C, Soliman M, Kapadia M, Ding Y, Payne K, Dovat S. Casein Kinase II (CK2), Glycogen Synthase Kinase-3 (GSK-3) and Ikaros mediated regulation of leukemia. Adv Biol Regul 2017. [PMID: 28623166 DOI: 10.1016/j.jbior.2017.06.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Signaling networks that regulate cellular proliferation often involve complex interactions between several signaling pathways. In this manuscript we review the crosstalk between the Casein Kinase II (CK2) and Glycogen Synthase Kinase-3 (GSK-3) pathways that plays a critical role in the regulation of cellular proliferation in leukemia. Both CK2 and GSK-3 are potential targets for anti-leukemia treatment. Previously published data suggest that CK2 and GSK-3 act synergistically to promote the phosphatidylinositol-3 kinase (PI3K) pathway via phosphorylation of PTEN. More recent data demonstrate another mechanism through which CK2 promotes the PI3K pathway - via transcriptional regulation of PI3K pathway genes by the newly-discovered CK2-Ikaros axis. Together, these data suggest that the CK2 and GSK-3 pathways regulate AKT/PI3K signaling in leukemia via two complementary mechanisms: a) direct phosphorylation of PTEN and b) transcriptional regulation of PI3K-promoting genes. Functional interactions between CK2, Ikaros and GSK3 define a novel signaling network that regulates proliferation of leukemia cells. This regulatory network involves both direct posttranslational modifications (by CK and GSK-3) and transcriptional regulation (via CK2-mediated phosphorylation of Ikaros). This information provides a basis for the development of targeted therapy for leukemia.
Collapse
Affiliation(s)
- Chandrika Gowda
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Mario Soliman
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Malika Kapadia
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Yali Ding
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Kimberly Payne
- Department of Anatomy, Loma Linda University, Loma Linda, CA, USA.
| | - Sinisa Dovat
- Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
164
|
Jeon KI, Phipps RP, Sime PJ, Huxlin KR. Antifibrotic Actions of Peroxisome Proliferator-Activated Receptor γ Ligands in Corneal Fibroblasts Are Mediated by β-Catenin-Regulated Pathways. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1660-1669. [PMID: 28606794 DOI: 10.1016/j.ajpath.2017.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 03/30/2017] [Accepted: 04/05/2017] [Indexed: 01/08/2023]
Abstract
Wound healing after corneal injury typically involves fibrosis, with transforming growth factor β1 (TGF-β1) as one of its strongest mediators. A class of small molecules-peroxisome proliferator-activated receptor γ (PPARγ) ligands-exert potent antifibrotic effects in the cornea by blocking phosphorylation of p38 mitogen-activated protein kinase (MAPK). However, why this blocks fibrosis remains unknown. Herein, we show that PPARγ ligands (rosiglitazone, troglitazone, and 15-deoxy-Δ12,14-prostaglandin J2) decrease levels of β-catenin. We also show that β-catenin siRNA and the Wingless/integrated (Wnt) inhibitor pyrvinium block the ability of corneal fibroblasts to up-regulate synthesis of α-smooth muscle actin (α-SMA), collagen 1 (COL1), and fibronectin (FN) in response to TGF-β1. Activation of TGF-β receptors and p38 MAPK increased glycogen synthase kinase 3β (GSK3β) phosphorylation, whereas a chemical inhibitor of p38 MAPK (SB203580) reduced the phosphorylation of GSK3β, decreasing active β-catenin levels in both cytoplasmic and nuclear fractions. Finally, lithium chloride, a GSK3 inhibitor, also attenuated the TGF-β1-induced increase in α-SMA, COL1, and FN expression. All in all, our results suggest that TGF-β1 stimulation increases active β-catenin concentration in cultured corneal fibroblasts through p38 MAPK regulation of canonical Wnt/β-catenin signaling, increasing α-SMA, COL1, and FN synthesis. Thus, PPARγ ligands, by blocking TGF-β1-induced p38 MAPK phosphorylation, prevent increases in both total and active β-catenin through p38 MAPK-GSK3β signaling.
Collapse
Affiliation(s)
- Kye-Im Jeon
- Flaum Eye Institute, University of Rochester, Rochester, New York
| | - Richard P Phipps
- Flaum Eye Institute, University of Rochester, Rochester, New York; Department of Medicine, University of Rochester, Rochester, New York; Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Patricia J Sime
- Department of Medicine, University of Rochester, Rochester, New York; Department of Environmental Medicine, University of Rochester, Rochester, New York
| | - Krystel R Huxlin
- Flaum Eye Institute, University of Rochester, Rochester, New York; Center for Visual Science, University of Rochester, Rochester, New York.
| |
Collapse
|
165
|
Bertrand JU, Petit V, Hacker E, Berlin I, Hayward NK, Pouteaux M, Sage E, Whiteman DC, Larue L. UVB represses melanocyte cell migration and acts through β-catenin. Exp Dermatol 2017; 26:875-882. [DOI: 10.1111/exd.13318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/08/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Juliette U. Bertrand
- Institut Curie; PSL Research University; INSERM U1021; Normal and Pathological Development of Melanocytes; Orsay France
- Univ Paris-Sud; Univ Paris-Saclay; CNRS UMR 3347; Orsay France
- Equipe Labellisée Ligue Contre le Cancer; Orsay France
| | - Valérie Petit
- Institut Curie; PSL Research University; INSERM U1021; Normal and Pathological Development of Melanocytes; Orsay France
- Univ Paris-Sud; Univ Paris-Saclay; CNRS UMR 3347; Orsay France
- Equipe Labellisée Ligue Contre le Cancer; Orsay France
| | - Elke Hacker
- Queensland Institute of Medical Research; Brisbane QLD Australia
| | - Irina Berlin
- Institut Curie; PSL Research University; INSERM U1021; Normal and Pathological Development of Melanocytes; Orsay France
- Univ Paris-Sud; Univ Paris-Saclay; CNRS UMR 3347; Orsay France
- Equipe Labellisée Ligue Contre le Cancer; Orsay France
| | | | - Marie Pouteaux
- Institut Curie; PSL Research University; INSERM U1021; Normal and Pathological Development of Melanocytes; Orsay France
- Univ Paris-Sud; Univ Paris-Saclay; CNRS UMR 3347; Orsay France
- Equipe Labellisée Ligue Contre le Cancer; Orsay France
| | - Evelyne Sage
- Institut Curie; PSL Research University; INSERM U1021; Normal and Pathological Development of Melanocytes; Orsay France
- Univ Paris-Sud; Univ Paris-Saclay; CNRS UMR 3347; Orsay France
- Equipe Labellisée Ligue Contre le Cancer; Orsay France
| | | | - Lionel Larue
- Institut Curie; PSL Research University; INSERM U1021; Normal and Pathological Development of Melanocytes; Orsay France
- Univ Paris-Sud; Univ Paris-Saclay; CNRS UMR 3347; Orsay France
- Equipe Labellisée Ligue Contre le Cancer; Orsay France
| |
Collapse
|
166
|
Kim H, Kwon KW, Kim WS, Shin SJ. Virulence-dependent induction of interleukin-10-producing-tolerogenic dendritic cells by Mycobacterium tuberculosis impedes optimal T helper type 1 proliferation. Immunology 2017; 151:177-190. [PMID: 28140445 DOI: 10.1111/imm.12721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/16/2017] [Accepted: 01/24/2017] [Indexed: 01/03/2023] Open
Abstract
Mycobacterium tuberculosis inhibits optimal T helper type 1 (Th1) responses during infection. However, the precise mechanisms by which virulent M. tuberculosis limits Th1 responses remain unclear. Here, we infected dendritic cells (DCs) with the virulent M. tuberculosis strain H37Rv or the attenuated strain H37Ra to investigate the phenotypic and functional alterations in DCs and resultant T-cell responses. H37Rv-infected DCs suppressed Th1 responses more strongly than H37Ra-infected DCs. Interestingly, H37Rv, but not H37Ra, impaired DC surface molecule expression (CD80, CD86 and MHC class II) due to prominent interleukin-10 (IL-10) production while augmenting the expression of tolerogenic molecules including PD-L1, CD103, Tim-3 and indoleamine 2,3-dioxygenase on DCs in a multiplicity-of-infection (MOI) -dependent manner. These results indicate that virulent M. tuberculosis drives immature DCs toward a tolerogenic phenotype. Notably, the tolerogenic phenotype of H37Rv-infected DCs was blocked in DCs generated from IL-10-/- mice or DCs treated with an IL-10-neutralizing monoclonal antibody, leading to restoration of Th1 polarization. These findings suggest that IL-10 induces a tolerogenic DC phenotype. Interestingly, p38 mitogen-activated protein kinase (MAPK) activation predominantly mediates IL-10 production; hence, H37Rv tends to induce a tolerogenic DC phenotype through expression of tolerogenic molecules in the p38 MAPK-IL-10 axis. Therefore, suppressing the tolerogenic cascade in DCs is a novel strategy for stimulating optimal protective T-cell responses against M. tuberculosis infection.
Collapse
Affiliation(s)
- Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Kee Woong Kwon
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Woo Sik Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
167
|
Abstract
Glycogen synthase kinase-3 (GSK-3) is a ubiquitously expressed protein kinase that sits at the nexus of multiple signaling pathways. Its deep integration into cellular control circuits is consummate to its implication in diseases ranging from mood disorders to diabetes to neurodegenerative diseases and cancers. The selectivity and insulation of such a promiscuous kinase from unwanted crosstalk between pathways, while orchestrating a multifaceted response to cellular stimuli, offer key insights into more general mechanisms of cell regulation. Here, we review recent advances that have contributed to the understanding of GSK-3 and its role in driving appreciation of intracellular signal coordination.
Collapse
Affiliation(s)
- Kevin W Cormier
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - James R Woodgett
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| |
Collapse
|
168
|
Inverse correlation between the metastasis suppressor RKIP and the metastasis inducer YY1: Contrasting roles in the regulation of chemo/immuno-resistance in cancer. Drug Resist Updat 2017; 30:28-38. [PMID: 28363333 DOI: 10.1016/j.drup.2017.01.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/28/2016] [Accepted: 01/04/2017] [Indexed: 02/06/2023]
Abstract
Several gene products have been postulated to mediate inherent and/or acquired anticancer drug resistance and tumor metastasis. Among these, the metastasis suppressor and chemo-immuno-sensitizing gene product, Raf Kinase Inhibitor Protein (RKIP), is poorly expressed in many cancers. In contrast, the metastasis inducer and chemo-immuno-resistant factor Yin Yang 1 (YY1) is overexpressed in many cancers. This inverse relationship between RKIP and YY1 expression suggests that these two gene products may be regulated via cross-talks of molecular signaling pathways, culminating in the expression of different phenotypes based on their targets. Analyses of the molecular regulation of the expression patterns of RKIP and YY1 as well as epigenetic, post-transcriptional, and post-translational regulation revealed the existence of several effector mechanisms and crosstalk pathways, of which five pathways of relevance have been identified and analyzed. The five examined cross-talk pathways include the following loops: RKIP/NF-κB/Snail/YY1, p38/MAPK/RKIP/GSK3β/Snail/YY1, RKIP/Smurf2/YY1/Snail, RKIP/MAPK/Myc/Let-7/HMGA2/Snail/YY1, as well as RKIP/GPCR/STAT3/miR-34/YY1. Each loop is comprised of multiple interactions and cascades that provide evidence for YY1's negative regulation of RKIP expression and vice versa. These loops elucidate potential prognostic motifs and targets for therapeutic intervention. Chiefly, these findings suggest that targeted inhibition of YY1 by specific small molecule inhibitors and/or the specific induction of RKIP expression and activity are potential therapeutic strategies to block tumor growth and metastasis in many cancers, as well as to overcome anticancer drug resistance. These strategies present potential alternatives for their synergistic uses in combination with low doses of conventional chemo-immunotherapeutics and hence, increasing survival, reducing toxicity, and improving quality of life.
Collapse
|
169
|
CD5-CK2 Signaling Modulates Erk Activation and Thymocyte Survival. PLoS One 2016; 11:e0168155. [PMID: 28030587 PMCID: PMC5193405 DOI: 10.1371/journal.pone.0168155] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 11/24/2016] [Indexed: 12/31/2022] Open
Abstract
CD5 is well recognized for its importance in thymic selection. Although this property of CD5 has been attributed to its ITIM-domain dependent regulation of TCR-signal strength, the mechanism has not been established. A second major signaling domain within the cytoplasmic tail of CD5 is a CK2 binding/activation domain (CD5-CK2BD). Using a gene-targeted mouse in which the CD5-CK2BD is selectively ablated (CD5-ΔCK2BD), we determined that loss of function of CD5-CK2 signaling in a MHC-II selecting TCR transgenic (OT-II) mouse resulted in decrease in double positive (DP) thymocytes, which correlated with enhanced apoptosis. Remarkably, DP cells expressing high levels of CD5 and CD69 and single positive (CD4+SP) thymocytes were increased in CD5-ΔCK2BD mice indicating that CD5-CK2 signaling regulates positive selection and promotes survival. Consistent with this possibility, we determined that the activation and nuclear localization of ERK as well as apoptosis was greater in thymic populations from OTII CD5-ΔCK2BD mice than OTII CD5-WT mice following injection of OVA323-339-peptide. The mobilization of Ca2+, an early event of TCR activation, was not altered by the loss of CD5-CK2 signaling. Collectively, these data demonstrate that the CD5-CK2 signaling axis regulates positive selection by modulating activation of ERK and promoting survival independent of proximal TCR signals.
Collapse
|
170
|
Kim JG, Kim MJ, Choi WJ, Moon MY, Kim HJ, Lee JY, Kim J, Kim SC, Kang SG, Seo GY, Kim PH, Park JB. Wnt3A Induces GSK-3β Phosphorylation and β-Catenin Accumulation Through RhoA/ROCK. J Cell Physiol 2016; 232:1104-1113. [DOI: 10.1002/jcp.25572] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/29/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Jae-Gyu Kim
- Department of Biochemistry; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
| | - Myoung-Ju Kim
- Department of Biochemistry; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
| | - Won-Ji Choi
- Department of Biochemistry; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
| | - Mi-Young Moon
- Department of Biochemistry; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
| | - Hee-Jun Kim
- Department of Biochemistry; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
| | - Jae-Yong Lee
- Department of Biochemistry; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
- Institute of Cell Differentiation and Aging; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
| | - Jaebong Kim
- Department of Biochemistry; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
- Institute of Cell Differentiation and Aging; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
| | - Sung-Chan Kim
- Department of Biochemistry; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
- Institute of Cell Differentiation and Aging; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
| | - Seung Goo Kang
- Division of Biomedical Convergence; School of Biomedical Science and Institute of Bioscience and Biotechnology; Kangwon National University; Chuncheon Kangwon-do Republic of Korea
| | - Goo-Young Seo
- Department of Molecular Bioscience; School of Biomedical Science and Institute of Bioscience and Biotechnology; Kangwon National University; Chuncheon Kangwon-do Republic of Korea
| | - Pyeung-Hyeun Kim
- Department of Molecular Bioscience; School of Biomedical Science and Institute of Bioscience and Biotechnology; Kangwon National University; Chuncheon Kangwon-do Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
- Institute of Cell Differentiation and Aging; Hallym University College of Medicine; Chuncheon Kangwon-do Republic of Korea
| |
Collapse
|
171
|
Grabinski T, Kanaan NM. Novel Non-phosphorylated Serine 9/21 GSK3β/α Antibodies: Expanding the Tools for Studying GSK3 Regulation. Front Mol Neurosci 2016; 9:123. [PMID: 27909397 PMCID: PMC5112268 DOI: 10.3389/fnmol.2016.00123] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 11/02/2016] [Indexed: 12/25/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) β and α are serine/threonine kinases involved in many biological processes. A primary mechanism of GSK3 activity regulation is phosphorylation of N-terminal serine (S) residues (S9 in GSK3β, S21 in GSK3α). Phosphorylation is inhibitory to GSK3 kinase activity because the phosphorylated N-terminus acts as a competitive inhibitor for primed substrates. Despite widespread interest in GSK3 across most fields of biology, the research community does not have reagents that specifically react with nonphosphoS9/21 GSK3β/α (the so-called "active" form). Here, we describe two novel monoclonal antibodies that specifically react with nonphosphoS9/21 GSK3β/α in multiple species (human, mouse, and rat). One of the antibodies is specific for nonphospho-S9 GSK3β (clone 12B2) and one for nonphospho-S9/21 GSK3β/α (clone 15C2). These reagents were validated for specificity and reactivity in several biochemical and immunochemical assays, and they show linear detection of nonphosphoS GSK3. Finally, these reagents provide significant advantages in studying GSK3β regulation. We used both antibodies to study the regulation of S9 phosphorylation by Akt and protein phosphatases. We used 12B2 (due to its specificity for GSK3β) and to demonstrate that protein phosphatase inhibition reduces nonphospho-S9 GSK3β levels and lowers kinase activity within cells. The ability to use the same reagent across biochemical, immunohistological and kinase activity assays provides a powerful approach for studying serine-dependent regulation of GSK3β/α.
Collapse
Affiliation(s)
- Tessa Grabinski
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand RapidsMI, USA
| | - Nicholas M. Kanaan
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand RapidsMI, USA
- Hauenstein Neuroscience Center, Mercy Health Saint Mary’s, Grand RapidsMI, USA
| |
Collapse
|
172
|
Sutariya B, Jhonsa D, Saraf MN. TGF-β: the connecting link between nephropathy and fibrosis. Immunopharmacol Immunotoxicol 2016; 38:39-49. [PMID: 26849902 DOI: 10.3109/08923973.2015.1127382] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Renal fibrosis is the usual outcome of an excessive accumulation of extracellular matrix (ECM) that frequently occurs in membranous and diabetic nephropathy. The result of renal fibrosis would be end-stage renal failure, which requires costly dialysis or kidney transplantation. Renal fibrosis typically results from chronic inflammation via production of several molecules, such as growth factors, angiogenic factors, fibrogenic cytokines, and proteinase. All of these factors can stimulate excessive accumulation of ECM components through epithelial to mesenchymal transition (EMT), which results in renal fibrosis. Among these, transforming growth factor-beta (TGF-β) is proposed to be the major regulator in inducing EMT. Besides ECM protein synthesis, TGF-β is involved in hypertrophy, proliferation, and apoptosis in renal cells. In particular, TGF-β is likely to be most potent and ubiquitous profibrotic factor acting through several intracellular signaling pathways including protein kinases and transcription factors. Factors that regulate TGF-β expression in renal cell include hyperglycemia, angiotensin II, advance glycation end products, complement activation (C5b-9), and oxidative stress. Over the past several years, the common understanding of the pathogenic factors that lead to renal fibrosis in nephropathy has improved considerably. This review will discuss the recent findings on the mechanisms and role of TGF-β in membranous and diabetic nephropathy.
Collapse
Affiliation(s)
- Brijesh Sutariya
- a Department of Pharmacology , Bombay College of Pharmacy , Mumbai , Maharashtra , India
| | - Dimple Jhonsa
- a Department of Pharmacology , Bombay College of Pharmacy , Mumbai , Maharashtra , India
| | - Madhusudan N Saraf
- a Department of Pharmacology , Bombay College of Pharmacy , Mumbai , Maharashtra , India
| |
Collapse
|
173
|
Wang Y, Wang Y, Zhong T, Guo J, Li L, Zhang H, Wang L. Transcriptional regulation of pig GYS1 gene by glycogen synthase kinase 3β (GSK3β). Mol Cell Biochem 2016; 424:203-208. [PMID: 27785702 DOI: 10.1007/s11010-016-2856-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 10/22/2016] [Indexed: 01/02/2023]
Abstract
Glycogen synthase kinase 3β (GSK3β) is a ubiquitous serine/threonine kinase and has important roles in glycogen metabolism biosynthesis. Studies have revealed that GSK3β can directly regulate the glycogen synthase activity, yet little is known about the regulation of GSK3β on GYS1 gene transcription. Here, we show that overexpression of GSK3β decreased the mRNA expression level of GYS1. Then we cloned approximately 1.5 kb of pig GYS1 gene promoter region, generated sequential deletion constructs, and evaluated their activity. A gradual increase of the promoter activity was seen with increasing length of the promoter sequence, reaching its highest activity to the sequence corresponding to nt -350 to +224, and then decreased. However, the activities of constructed promoter fragments show different responses to GSK3β co-transfection. By analyzing a series of GYS1 promoter reporter constructs, we have defined two crucial regions (-1488 to -539, -350 to -147) that are responsible for GSK3β-induced transcriptional repression. Furthermore, the ChIP results revealed that only the first and second NF-κB sites of GYS1 promoter could bind to p65, and overexpression of GSK3β induced a significant decrease in p65 binding to the second NF-κB binding site, suggesting that GSK3β may regulate expression of GYS1 gene through binding to the second rather than the first NF-κB site. These data suggest that the NF-κB plays important roles in the transcriptional activity of pig GYS1 gene regulated by GSK3β.
Collapse
Affiliation(s)
- Yilin Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China
| | - Yan Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China
| | - Tao Zhong
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China
| | - Jiazhong Guo
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China
| | - Li Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China
| | - Hongping Zhang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China.,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China
| | - Linjie Wang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China. .,Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
174
|
Gu M, Liu Z, Lai R, Liu S, Lin W, Ouyang C, Ye S, Huang H, Wang X. RKIP and TBK1 form a positive feedback loop to promote type I interferon production in innate immunity. EMBO J 2016; 35:2553-2565. [PMID: 27753621 DOI: 10.15252/embj.201694060] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 09/14/2016] [Accepted: 09/19/2016] [Indexed: 11/09/2022] Open
Abstract
TANK-binding kinase 1 (TBK1) activation is a central event in type I interferon production in anti-virus innate immunity. However, the regulatory mechanism underlying TBK1 activation remains unclear. Here we report that Raf kinase inhibitory protein (RKIP) is essential for TBK1 activation and type I interferon production triggered by viral infection. Upon viral infection, RKIP is phosphorylated at serine 109 (S109) by TBK1. Phosphorylation of RKIP enhances its interaction with TBK1 and in turn promotes TBK1 autophosphorylation. Mutation of RKIP S109 to alanine abrogates the interaction between RKIP and TBK1, and the anti-viral function of RKIP RKIP deficiency inhibits intracellular double-stranded RNA- or DNA-induced type I interferon production. Consistently, RKIP deficiency renders the mice more susceptible to vesicular stomatitis virus (VSV) and herpes simplex virus (HSV) infections. This study reveals a previously unrecognized positive feedback loop between RKIP and TBK1 that is essential for type I interferon production in anti-viral innate immunity.
Collapse
Affiliation(s)
- Meidi Gu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhiyong Liu
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Rongrong Lai
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Si Liu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - Wenlong Lin
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chuan Ouyang
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Ye
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaojian Wang
- Institute of Immunology, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
175
|
Wong SY, Tan MGK, Wong PTH, Herr DR, Lai MKP. Andrographolide induces Nrf2 and heme oxygenase 1 in astrocytes by activating p38 MAPK and ERK. J Neuroinflammation 2016; 13:251. [PMID: 27663973 PMCID: PMC5034653 DOI: 10.1186/s12974-016-0723-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/15/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Andrographolide is the major labdane diterpenoid originally isolated from Andrographis paniculata and has been shown to have anti-inflammatory and antioxidative effects. However, there is a dearth of studies on the potential therapeutic utility of andrographolide in neuroinflammatory conditions. Here, we aimed to investigate the mechanisms underlying andrographolide's effect on the expression of anti-inflammatory and antioxidant heme oxygenase-1 (HO-1) in primary astrocytes. METHODS Measurements of the effects of andrograholide on antioxidant HO-1 and its transcription factor, Nrf2, include gene expression, protein turnover, and activation of putative signaling regulators. RESULTS Andrographolide potently activated Nrf2 and also upregulated HO-1 expression in primary astrocytes. Andrographolide's effects on Nrf2 seemed to be biphasic, with acute (within 1 h) reductions in Nrf2 ubiquitination efficiency and turnover rate, followed by upregulation of Nrf2 mRNA between 8 and 24 h. The acute regulation of Nrf2 by andrographolide seemed to be independent of Keap1 and partly mediated by p38 MAPK and ERK signaling. CONCLUSIONS These data provide further insights into the mechanisms underlying andrographolide's effects on astrocyte-mediated antioxidant, and anti-inflammatory responses and support the further assessment of andrographolide as a potential therapeutic for neurological conditions in which oxidative stress and neuroinflammation are implicated.
Collapse
Affiliation(s)
- Siew Ying Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, 117599 Singapore
| | - Michelle G. K. Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, 117599 Singapore
- Department of Clinical Research, Singapore General Hospital, Outram, Singapore
| | - Peter T. H. Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, 117599 Singapore
| | - Deron R. Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, 117599 Singapore
| | - Mitchell K. P. Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Unit 09-01, Centre for Translational Medicine (MD6), 14 Medical Drive, Kent Ridge, 117599 Singapore
| |
Collapse
|
176
|
McCubrey JA, Rakus D, Gizak A, Steelman LS, Abrams SL, Lertpiriyapong K, Fitzgerald TL, Yang LV, Montalto G, Cervello M, Libra M, Nicoletti F, Scalisi A, Torino F, Fenga C, Neri LM, Marmiroli S, Cocco L, Martelli AM. Effects of mutations in Wnt/β-catenin, hedgehog, Notch and PI3K pathways on GSK-3 activity-Diverse effects on cell growth, metabolism and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2942-2976. [PMID: 27612668 DOI: 10.1016/j.bbamcr.2016.09.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 02/07/2023]
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that participates in an array of critical cellular processes. GSK-3 was first characterized as an enzyme that phosphorylated and inactivated glycogen synthase. However, subsequent studies have revealed that this moon-lighting protein is involved in numerous signaling pathways that regulate not only metabolism but also have roles in: apoptosis, cell cycle progression, cell renewal, differentiation, embryogenesis, migration, regulation of gene transcription, stem cell biology and survival. In this review, we will discuss the roles that GSK-3 plays in various diseases as well as how this pivotal kinase interacts with multiple signaling pathways such as: PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, Wnt/beta-catenin, hedgehog, Notch and TP53. Mutations that occur in these and other pathways can alter the effects that natural GSK-3 activity has on regulating these signaling circuits that can lead to cancer as well as other diseases. The novel roles that microRNAs play in regulation of the effects of GSK-3 will also be evaluated. Targeting GSK-3 and these other pathways may improve therapy and overcome therapeutic resistance.
Collapse
Affiliation(s)
- James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA.
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Agnieszka Gizak
- Department of Animal Molecular Physiology, Institute of Experimental Biology, Wroclaw University, Wroclaw, Poland
| | - Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Steve L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University Greenville, NC 27858, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Timothy L Fitzgerald
- Department of Surgery, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Massimo Libra
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | - Aurora Scalisi
- Unit of Oncologic Diseases, ASP-Catania, Catania 95100, Italy
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Concettina Fenga
- Department of Biomedical, Odontoiatric, Morphological and Functional Images, Occupational Medicine Section - Policlinico "G. Martino" - University of Messina, Messina 98125, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Sandra Marmiroli
- Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| |
Collapse
|
177
|
Xanthatin anti-tumor cytotoxicity is mediated via glycogen synthase kinase-3β and β-catenin. Biochem Pharmacol 2016; 115:18-27. [DOI: 10.1016/j.bcp.2016.06.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 06/15/2016] [Indexed: 12/13/2022]
|
178
|
Dema A, Schröter MF, Perets E, Skroblin P, Moutty MC, Deàk VA, Birchmeier W, Klussmann E. The A-Kinase Anchoring Protein (AKAP) Glycogen Synthase Kinase 3β Interaction Protein (GSKIP) Regulates β-Catenin through Its Interactions with Both Protein Kinase A (PKA) and GSK3β. J Biol Chem 2016; 291:19618-30. [PMID: 27484798 DOI: 10.1074/jbc.m116.738047] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Indexed: 01/24/2023] Open
Abstract
The A-kinase anchoring protein (AKAP) GSK3β interaction protein (GSKIP) is a cytosolic scaffolding protein binding protein kinase A (PKA) and glycogen synthase kinase 3β (GSK3β). Here we show that both the AKAP function of GSKIP, i.e. its direct interaction with PKA, and its direct interaction with GSK3β are required for the regulation of β-catenin and thus Wnt signaling. A cytoplasmic destruction complex targets β-catenin for degradation and thus prevents Wnt signaling. Wnt signals cause β-catenin accumulation and translocation into the nucleus, where it induces Wnt target gene expression. GSKIP facilitates control of the β-catenin stabilizing phosphorylation at Ser-675 by PKA. Its interaction with GSK3β facilitates control of the destabilizing phosphorylation of β-catenin at Ser-33/Ser-37/Thr-41. The influence of GSKIP on β-catenin is explained by its scavenger function; it recruits the kinases away from the destruction complex without forming a complex with β-catenin. The regulation of β-catenin by GSKIP is specific for this AKAP as AKAP220, which also binds PKA and GSK3β, did not affect Wnt signaling. We find that the binding domain of AKAP220 for GSK3β is a conserved GSK3β interaction domain (GID), which is also present in GSKIP. Our findings highlight an essential compartmentalization of both PKA and GSK3β by GSKIP, and ascribe a function to a cytosolic AKAP-PKA interaction as a regulatory factor in the control of canonical Wnt signaling. Wnt signaling controls different biological processes, including embryonic development, cell cycle progression, glycogen metabolism, and immune regulation; deregulation is associated with diseases such as cancer, type 2 diabetes, inflammatory, and Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
- Alessandro Dema
- From the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany and
| | - Micha Friedemann Schröter
- From the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany and
| | - Ekaterina Perets
- From the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany and
| | - Philipp Skroblin
- From the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany and
| | - Marie Christine Moutty
- From the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany and
| | - Veronika Anita Deàk
- From the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany and
| | - Walter Birchmeier
- From the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany and
| | - Enno Klussmann
- From the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Strasse 10, 13125 Berlin, Germany and the DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Oudenarder Strasse 16, 13347 Berlin, Germany
| |
Collapse
|
179
|
Sakisaka Y, Kanaya S, Nakamura T, Tamura M, Shimauchi H, Nemoto E. p38 MAP kinase is required for Wnt3a-mediated osterix expression independently of Wnt-LRP5/6-GSK3β signaling axis in dental follicle cells. Biochem Biophys Res Commun 2016; 478:527-32. [PMID: 27450807 DOI: 10.1016/j.bbrc.2016.07.076] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 07/18/2016] [Indexed: 11/24/2022]
Abstract
Wnt3a is a secreted glycoprotein that activates the glycogen synthase kinase-3β (GSK3β)/β-catenin signaling pathway through low-density-lipoprotein receptor-related protein (LRP)5/6 co-receptors. Wnt3a has been implicated in periodontal development and homeostasis, as well as in cementum formation. Recently, we have reported that Wnt3a increases alkaline phosphatase expression through the induction of osterix (Osx) expression in dental follicle cells, a precursor of cementoblasts. However, the molecular mechanism by which Wnt3a induces Osx expression is still unknown. In this study, we show that Wnt3a-induced Osx expression was inhibited in the presence of p38 mitogen-activated protein kinase (MAPK) inhibitors (SB203580 and SB202190) at gene and protein levels, as assessed by real-time PCR and immunocytohistochemistry, respectively. Pretreatment of cells with Dickkopf-1, a potent canonical Wnt antagonist binding to LRP5/6 co-receptors, did not influence Wnt3a-mediated p38 MAPK phosphorylation, suggesting that Wnt3a activates p38 MAPK through LRP5/6-independent signaling. On the other hand, pretreatment with p38 MAPK inhibitors had no effects on the phosphorylated status of GSK3β and β-catenin as well as β-catenin nuclear translocation, but inhibited Wnt3a-mediated β-catenin transcriptional activity. These findings suggest that p38 MAPK modulates canonical Wnt signaling at the β-catenin transcriptional level without any crosstalk with the Wnt3a-mediated LRP5/6-GSK3β signaling axis and subsequent β-catenin nuclear translocation. These findings expand our knowledge of the mechanisms controlling periodontal development and regeneration.
Collapse
Affiliation(s)
- Yukihiko Sakisaka
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Sousuke Kanaya
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan; Liason Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Takashi Nakamura
- Department of Dental Pharmacology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Masato Tamura
- Department of Biochemistry and Molecular Biology, Hokkaido University Graduate School of Dentistry, Sapporo 060-8586, Japan
| | - Hidetoshi Shimauchi
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan
| | - Eiji Nemoto
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai 980-8575, Japan.
| |
Collapse
|
180
|
Jiang GY, Zhang XP, Zhang Y, Xu HT, Wang L, Li QC, Wang EH. Coiled-coil domain-containing protein 8 inhibits the invasiveness and migration of non-small cell lung cancer cells. Hum Pathol 2016; 56:64-73. [PMID: 27342910 DOI: 10.1016/j.humpath.2016.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/03/2016] [Accepted: 06/11/2016] [Indexed: 01/08/2023]
Abstract
Lung cancer has always been the leading cause of death among patients with malignant tumors, and the majority of these patients die because of cancer cell invasion and metastasis. Previous studies have implicated coiled-coil domain-containing protein 8 (CCDC8) as a tumor suppressor in several types of cancer, such as breast and prostate cancers. However, the expression levels or functions of CCDC8 in lung cancer have not been elucidated. Here, we used immunohistochemical staining to measure CCDC8 expression in 147 samples from tumors and 30 samples from the adjacent normal lung tissues of patients with non-small cell lung cancer. CCDC8 was shown to be located predominantly in the cytoplasm and partially on the cell membrane, and its expression level was significantly lower in lung cancer samples than that in the adjacent normal lung tissues (P=.001). CCDC8 expression was closely related to tumor differentiation (P=.039), tumor-node-metastasis stage (P=.009), lymph node metastasis (P=.038), and prognosis (P=.043) of lung cancer. Transfection of A549 cells with CCDC8 significantly reduced cell invasion and migration (P<.05), whereas the invasiveness and migration capacity in CCDC8-knockdown A549 cells were significantly increased in comparison with the control cells (P<.05). Furthermore, we demonstrated that CCDC8 can downregulate the expression of Snail and upregulate the expression of E-cadherin by inhibiting p-P38 and p-IκBα. Collectively, CCDC8 may suppress the invasion and metastasis of lung cancer cells, and it may represent a promising therapeutic target for non-small cell lung cancer.
Collapse
Affiliation(s)
- Gui-Yang Jiang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, China
| | - Xiu-Peng Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, China
| | - Yong Zhang
- Department of Pathology, Cancer Hospital of China Medical University, Shenyang 110042, China
| | - Hong-Tao Xu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, China
| | - Liang Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, China
| | - Qing-Chang Li
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, China.
| | - En-Hua Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang 110001, China
| |
Collapse
|
181
|
Abstract
Fibrosis occurs in systemic tissues other than the brain and finally induces dysfunction of the fibrotic organ. Kidney fibrosis is related to scarring after acute kidney injury and the progression of chronic kidney disease. Kidney function decreases with the progression of kidney fibrosis. As fibrotic tissue cannot return to its original status, advanced kidney fibrosis requires the administration of dialysis or kidney transplantation. Thus, elucidation the mechanism of kidney fibrosis is an important research theme. The proliferation and activation of (myo) fibroblasts and the excessive production of an extracellular matrix are common mechanisms in fibrosis in many organs, but it seems that kidney fibrosis has specific pathways. Tubular epithelial, mesangial cells, and erythropoietin producing cells, which exist only in the kidney, participate in forming kidney fibrosis. This review highlights an understanding of the cells and their underlying mechanisms, which are specific to kidney fibrosis process: transforming growth factor-β (TGF-β), epithelial-mesenchymal transition, wingless/int-1 (WNT) signaling, renal anemia, and uremia. Finally, we describe potential therapies that focus on the mechanisms of kidney fibrosis: anti-TGF-β antibody and mammalian target of rapamycin (mTOR).
Collapse
|
182
|
Osei-Sarfo K, Urvalek AM, Tang XH, Scognamiglio T, Gudas LJ. Initiation of esophageal squamous cell carcinoma (ESCC) in a murine 4-nitroquinoline-1-oxide and alcohol carcinogenesis model. Oncotarget 2016; 6:6040-52. [PMID: 25714027 PMCID: PMC4467420 DOI: 10.18632/oncotarget.3339] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/04/2015] [Indexed: 01/14/2023] Open
Abstract
Esophageal squamous cell carcinomas (ESCCs) are very common, aggressive tumors, and are often associated with alcohol and tobacco abuse. Because ESCCs exhibit high recurrence rates and are diagnosed at late stages, identification of prognostic and drug targets for prevention and treatment is critical. We used the 4-nitroquinoline-1-oxide (4-NQO) murine model of oral carcinogenesis and the Meadows-Cook model of alcohol abuse to assess changes in the expression of molecular markers during the initial stages of ESCC. Combining these two models, which mimic chronic alcohol and tobacco abuse in humans, we detected increased cellular proliferation (EGFR and Ki67 expression), increased canonical Wnt signaling and downstream elements (β-catenin, FoxM1, and S100a4 protein levels), changes in cellular adhesive properties (reduced E-cadherin in the basal layer of the esophageal epithelium), and increased levels of phosphorylated ERK1/2 and p38. Additionally, we found that treatment with ethanol alone increased the numbers of epithelial cells expressing solute carrier family 2 (facilitated glucose transporter, member 1) (SLC2A1) and carbonic anhydrase IX (CAIX), and increased the phosphorylation of p38. Thus, we identified both 4-NQO- and ethanol-specific targets in the initial stages of esophageal carcinogenesis, which should lead to the development of potential markers and therapeutic targets for human ESCC.
Collapse
Affiliation(s)
- Kwame Osei-Sarfo
- Department of Pharmacology, Weill Cornell Medical College, New York, USA
| | - Alison M Urvalek
- Department of Pharmacology, Weill Cornell Medical College, New York, USA
| | - Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medical College, New York, USA
| | | | - Lorraine J Gudas
- Department of Pharmacology, Weill Cornell Medical College, New York, USA.,The Meyer Cancer Center, Weill Cornell Medical College, New York, USA
| |
Collapse
|
183
|
Ahuja S, Dogra D, Stainier DYR, Reischauer S. Id4 functions downstream of Bmp signaling to restrict TCF function in endocardial cells during atrioventricular valve development. Dev Biol 2016; 412:71-82. [PMID: 26892463 DOI: 10.1016/j.ydbio.2016.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/20/2016] [Accepted: 02/05/2016] [Indexed: 10/22/2022]
Abstract
The atrioventricular canal (AVC) connects the atrial and ventricular chambers of the heart and its formation is critical for the development of the cardiac valves, chamber septation and formation of the cardiac conduction system. Consequently, problems in AVC formation can lead to congenital defects ranging from cardiac arrhythmia to incomplete cardiac septation. While our knowledge about early heart tube formation is relatively comprehensive, much remains to be investigated about the genes that regulate AVC formation. Here we identify a new role for the basic helix-loop-helix factor Id4 in zebrafish AVC valve development and function. id4 is first expressed in the AVC endocardium and later becomes more highly expressed in the atrial chamber. TALEN induced inactivation of id4 causes retrograde blood flow at the AV canal under heat induced stress conditions, indicating defects in AV valve function. At the molecular level, we found that id4 inactivation causes misexpression of several genes important for AVC and AV valve formation including bmp4 and spp1. We further show that id4 appears to control the number of endocardial cells that contribute to the AV valves by regulating Wnt signaling in the developing AVC endocardium.
Collapse
Affiliation(s)
- Suchit Ahuja
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Deepika Dogra
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Didier Y R Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.
| |
Collapse
|
184
|
Maiden SL, Petrova YI, Gumbiner BM. Microtubules Inhibit E-Cadherin Adhesive Activity by Maintaining Phosphorylated p120-Catenin in a Colon Carcinoma Cell Model. PLoS One 2016; 11:e0148574. [PMID: 26845024 PMCID: PMC4742228 DOI: 10.1371/journal.pone.0148574] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 01/19/2016] [Indexed: 01/06/2023] Open
Abstract
Tight regulation of cadherin-mediated intercellular adhesions is critical to both tissue morphogenesis during development and tissue homeostasis in adults. Cell surface expression of the cadherin-catenin complex is often directly correlated with the level of adhesion, however, examples exist where cadherin appears to be inactive and cells are completely non-adhesive. The state of p120-catenin phosphorylation has been implicated in regulating the adhesive activity of E-cadherin but the mechanism is currently unclear. We have found that destabilization of the microtubule cytoskeleton, independent of microtubule plus-end dynamics, dephosphorylates p120-catenin and activates E-cadherin adhesion in Colo 205 cells. Through chemical screening, we have also identified several kinases as potential regulators of E-cadherin adhesive activity. Analysis of several p120-catenin phosphomutants suggests that gross dephosphorylation of p120-catenin rather than that of specific amino acids may trigger E-cadherin adhesion. Uncoupling p120-catenin binding to E-cadherin at the membrane causes constitutive adhesion in Colo 205 cells, further supporting an inhibitory role of phosphorylated p120-catenin on E-cadherin activity.
Collapse
Affiliation(s)
- Stephanie L. Maiden
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Department of Biology, Truman State University, Kirksville, Missouri, United States of America
| | - Yuliya I. Petrova
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
| | - Barry M. Gumbiner
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, Virginia, United States of America
- Seattle Children’s Research Institute and University of Washington School of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
185
|
Zhou YX, Shi Z, Singh P, Yin H, Yu YN, Li L, Walsh MP, Gui Y, Zheng XL. Potential Role of Glycogen Synthase Kinase-3β in Regulation of Myocardin Activity in Human Vascular Smooth Muscle Cells. J Cell Physiol 2016; 231:393-402. [PMID: 26129946 DOI: 10.1002/jcp.25084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 06/26/2015] [Indexed: 01/13/2023]
Abstract
Glycogen synthase kinase (GSK)-3β, a serine/threonine kinase with an inhibitory role in glycogen synthesis in hepatocytes and skeletal muscle, is also expressed in cardiac and smooth muscles. Inhibition of GSK-3β results in cardiac hypertrophy through reducing phosphorylation and increasing transcriptional activity of myocardin, a transcriptional co-activator for serum response factor. Myocardin plays critical roles in differentiation of smooth muscle cells (SMCs). This study, therefore, aimed to examine whether and how inhibition of GSK-3β regulates myocardin activity in human vascular SMCs. Treatment of SMCs with the GSK-3β inhibitors AR-A014418 and TWS 119 significantly reduced endogenous myocardin activity, as indicated by lower expression of myocardin target genes (and gene products), CNN1 (calponin), TAGLN1 (SM22), and ACTA2 (SM α-actin). In human SMCs overexpressing myocardin through the T-REx system, treatment with either GSK-3β inhibitor also inhibited the expression of CNN1, TAGLN1, and ACTA2. These effects of GSK-3β inhibitors were mimicked by transfection with GSK-3β siRNA. Notably, both AR-A014418 and TWS 119 decreased the serine/threonine phosphorylation of myocardin. The chromatin immunoprecipitation assay showed that AR-A014418 treatment reduced myocardin occupancy of the promoter of the myocardin target gene ACTA2. Overexpression of a dominant-negative GSK-3β mutant in myocardin-overexpressing SMCs reduced the expression of calponin, SM22, and SM α-actin. As expected, overexpression of constitutively active or wild-type GSK-3β in SMCs without myocardin overexpression increased expression of these proteins. In summary, our results indicate that inhibition of GSK-3β reduces myocardin transcriptional activity, suggesting a role for GSK-3β in myocardin transcriptional activity and smooth muscle differentiation.
Collapse
Affiliation(s)
- Yi-Xia Zhou
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Zhan Shi
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Pavneet Singh
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Hao Yin
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yan-Ni Yu
- Guiyang Medical University, Guizhou, China
| | - Long Li
- Guiyang Medical University, Guizhou, China
| | - Michael P Walsh
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yu Gui
- Department of Physiology and Pharmacology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
186
|
Thornton TM, Delgado P, Chen L, Salas B, Krementsov D, Fernandez M, Vernia S, Davis RJ, Heimann R, Teuscher C, Krangel MS, Ramiro AR, Rincón M. Inactivation of nuclear GSK3β by Ser(389) phosphorylation promotes lymphocyte fitness during DNA double-strand break response. Nat Commun 2016; 7:10553. [PMID: 26822034 PMCID: PMC4740185 DOI: 10.1038/ncomms10553] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/28/2015] [Indexed: 12/16/2022] Open
Abstract
Variable, diversity and joining (V(D)J) recombination and immunoglobulin class switch recombination (CSR) are key processes in adaptive immune responses that naturally generate DNA double-strand breaks (DSBs) and trigger a DNA repair response. It is unclear whether this response is associated with distinct survival signals that protect T and B cells. Glycogen synthase kinase 3β (GSK3β) is a constitutively active kinase known to promote cell death. Here we show that phosphorylation of GSK3β on Ser(389) by p38 MAPK (mitogen-activated protein kinase) is induced selectively by DSBs through ATM (ataxia telangiectasia mutated) as a unique mechanism to attenuate the activity of nuclear GSK3β and promote survival of cells undergoing DSBs. Inability to inactivate GSK3β through Ser(389) phosphorylation in Ser(389)Ala knockin mice causes a decrease in the fitness of cells undergoing V(D)J recombination and CSR. Preselection-Tcrβ repertoire is impaired and antigen-specific IgG antibody responses following immunization are blunted in Ser(389)GSK3β knockin mice. Thus, GSK3β emerges as an important modulator of the adaptive immune response.
Collapse
Affiliation(s)
- Tina M. Thornton
- Department of Medicine/Immunobiology, University of Vermont, Burlington, Vermont 05405, USA
| | - Pilar Delgado
- B Cell Biology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 328029, Spain
| | - Liang Chen
- Department of Immunology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Beatriz Salas
- Department of Medicine/Immunobiology, University of Vermont, Burlington, Vermont 05405, USA
| | - Dimitry Krementsov
- Department of Medicine/Immunobiology, University of Vermont, Burlington, Vermont 05405, USA
| | - Miriam Fernandez
- Department of Medicine/Immunobiology, University of Vermont, Burlington, Vermont 05405, USA
| | - Santiago Vernia
- Program in Molecular Medicine, University of Massachusetts, Worcester, Massachusetts 01605, USA
| | - Roger J. Davis
- Program in Molecular Medicine, University of Massachusetts, Worcester, Massachusetts 01605, USA
- Howard Hughes Medical Institute, Worcester, Massachusetts 01605, USA
| | - Ruth Heimann
- Department of Medicine/Radiology, University of Vermont, Burlington, Vermont 05405, USA
| | - Cory Teuscher
- Department of Medicine/Immunobiology, University of Vermont, Burlington, Vermont 05405, USA
| | - Michael S. Krangel
- Department of Immunology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Almudena R. Ramiro
- B Cell Biology Lab, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 328029, Spain
| | - Mercedes Rincón
- Department of Medicine/Immunobiology, University of Vermont, Burlington, Vermont 05405, USA
| |
Collapse
|
187
|
Li YH, Li YY, Fan GW, Yu JH, Duan ZZ, Wang LY, Yu B. Cardioprotection of ginsenoside Rb1 against ischemia/reperfusion injury is associated with mitochondrial permeability transition pore opening inhibition. Chin J Integr Med 2016:10.1007/s11655-015-2433-6. [PMID: 26740222 DOI: 10.1007/s11655-015-2433-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To investigate the role of ginsenoside Rb1 (Gs-Rb1) in cardioprotection against ischemia/reperfusion (I/R) or hypoxia/reoxygenation (H/R) injury and to explore whether the cardioprotective action is mediated via attenuating the formation of mitochondrial permeability transition pore (mPTP). METHODS A Langendorff-perfused model of rat heart was employed. I/R injury was induced by breaking off perfusion for 40 min then reperfusion for 60 min. Gs-Rb1 (100 μmol/L) were administrated for 10 min before I/R. Infarct size was estimated by the 2,3,5-triphenyl tetrazolium chloride (TTC) staining. Lactate dehydrogenase (LDH) and creatine kinase (CK) released from effluents were measured. Transmission electron microscopy was performed to assess morphological difference between cardiac mitochondrial isolated from I/R rats and Gs-Rb1 pretreated rats. Western blot analysis was used to determine phosphorylation of protein kinase B/Akt, and its downstream target glycogen synthase kinase 3β (GSK-3β). Incubation isolated cardiac mitochondria with Gs-Rb1, Ca2+-induced opening of the mPTP was assessed by the reduction of absorbance at 520 nm (A520). Neonatal rat cardiomyocytes were subjected to hypoxia 9 h followed by reoxygenation 4 h to induce H/R injury. After pretreated with different concentration of Gs-Rb1 (6.25, 25, 100 μmol/L ), cell viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5- diphenyltetrazolium bromide (MTT) method. The membrane potential was estimated by Rh123 fluorescence. mPTP opening was measured using the probe calcein-AM. RESULTS Gs-Rb1 100 μmol/L significantly reduced the infarct size of hearts (26.39%±11.67% vs. I/R group 56.68%±5.88%, P<0.01). Compared with the I/R group, Gs-Rb1 pretreatment decreased LDH and CK levels in the coronary effluent (P<0.05 or P<0.01) as well as attenuated destructive ultrastructure induced by I/R. The protective effect of Gs-Rb1 involved in phosphorylating protein kinase B/PKB (Akt) and GSK-3β. In mitochondria isolated from rat hearts, significant inhibition of Ca2+-induced swelling was observed in samples that were pretreated with Gs-Rb1 (6.25, 25, 100, 400 μmol/L) for 10 min. When cardiomyocytes were isolated from neonatal rat and subjected to H/R, cell viability was increased with treatment of Gs-Rb1 (6.25, 25, 100 μmol/L ). Gs-Rb1 inhibited mPTP opening and restored subsequent loss of mitochondrial membrane potential. CONCLUSION Gs-Rb1 presents cardioprotective effect against I/R or H/R injury which involves in activating Akt, phosphorylating GSK-3β and inhibiting mPTP opening.
Collapse
Affiliation(s)
- Yu-Hong Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yan-Yan Li
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Guan-Wei Fan
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Jia-Hui Yu
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Zhen-Zhen Duan
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ling-Yan Wang
- State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- State Key Laboratory of Modern Chinese Medicine, Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Bin Yu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
188
|
Xing HY, Cai YQ, Wang XF, Wang LL, Li P, Wang GY, Chen JH. The Cytoprotective Effect of Hyperoside against Oxidative Stress Is Mediated by the Nrf2-ARE Signaling Pathway through GSK-3β Inactivation. PLoS One 2015; 10:e0145183. [PMID: 26674355 PMCID: PMC4682950 DOI: 10.1371/journal.pone.0145183] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 11/30/2015] [Indexed: 12/21/2022] Open
Abstract
Glycogen synthase kinase-3β (GSK-3β) acts as a negative regulator of NF-E2 related factor 2 (Nrf2) by inducing Nrf2 degradation and nuclear export. Our previous study demonstrated that the flavonoid hyperoside elicits cytoprotection against oxidative stress by activating the Keap1-Nrf2-ARE signaling pathway, thus increasing the expression of antioxidant enzymes, such as heme oxygenase-1 (HO-1), superoxide dismutase (SOD) and catalase. However, the role of GSK-3β in hyperoside-mediated Nrf2 activation is unclear. Here, we demonstrate that in a normal human hepatocyte cell line, (L02), hyperoside is capable of inducing the phosphorylation of GSK-3β at Ser9 without affecting the protein levels of GSK-3β and its phosphorylation at Thr390. Lithium chloride (LiCl) and short interfering RNA (siRNA)-mediated inhibition of GSK-3β significantly enhanced the ability of hyperoside to protect L02 liver cells from H2O2-induced oxidative damage, leading to increased cell survival shown by the maintenance of cell membrane integrity and elevated levels of glutathione (GSH), one of the endogenous antioxidant biomarkers. Further study showed that LiCl and siRNA-mediated inhibition of GSK-3β increased hyperoside-induced HO-1 expression, and the effect was dependent upon enhanced Nrf2 nuclear translocation and gene expression. These activities were followed by ARE-mediated transcriptional activation in the presence of hyperoside, which was abolished by the transfection of the cells with Nrf2 siRNA. Furthermore, the siRNA-mediated inhibition of Keap1 also enhanced hyperoside-induced Nrf2 nuclear accumulation and HO-1 expression, which was relatively smaller than the effects obtained from GSK-3β siRNA administration. Moreover, Keap1 siRNA administration alone had no significant effect on the phosphorylation and protein expression of GSK-3β. Collectively, our data provide evidence that hyperoside attenuates H2O2 -induced L02 cell damage by activating the Nrf2-ARE signaling pathway through both an increase in GSK-3β inhibitory phosphorylation at Ser9 and an inhibition of Keap1 and that hyperoside-mediated GSK-3β inhibition exhibits more significant effects.
Collapse
Affiliation(s)
- Hai-Yan Xing
- Department of Pharmacy, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Yong-Qing Cai
- Department of Pharmacy, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Xian-Feng Wang
- Department of Pharmacy, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Lin-Li Wang
- Department of Pharmacy, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Pan Li
- Department of Pharmacy, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Guan-Ying Wang
- Department of Pharmacy, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
| | - Jian-Hong Chen
- Department of Pharmacy, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing 400042, China
- * E-mail:
| |
Collapse
|
189
|
Prolyl isomerase Pin1 regulates cadmium-induced autophagy via ubiquitin-mediated post-translational stabilization of phospho-Ser GSK3αβ in human hepatocellular carcinoma cells. Biochem Pharmacol 2015; 98:511-21. [DOI: 10.1016/j.bcp.2015.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/08/2015] [Indexed: 11/22/2022]
|
190
|
Chen L, Yang J, Huang T, Kong X, Lu L, Cai YD. Mining for novel tumor suppressor genes using a shortest path approach. J Biomol Struct Dyn 2015. [PMID: 26209080 DOI: 10.1080/07391102.2015.1042915] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cancer, being among the most serious diseases, causes many deaths every year. Many investigators have devoted themselves to designing effective treatments for this disease. Cancer always involves abnormal cell growth with the potential to invade or spread to other parts of the body. In contrast, tumor suppressor genes (TSGs) act as guardians to prevent a disordered cell cycle and genomic instability in normal cells. Studies on TSGs can assist in the design of effective treatments against cancer. In this study, we propose a computational method to discover potential TSGs. Based on the known TSGs, a number of candidate genes were selected by applying the shortest path approach in a weighted graph that was constructed using protein-protein interaction network. The analysis of selected genes shows that some of them are new TSGs recently reported in the literature, while others may be novel TSGs.
Collapse
Affiliation(s)
- Lei Chen
- a College of Life Science , Shanghai University , Shanghai 200444 , P.R. China.,b College of Information Engineering , Shanghai Maritime University , Shanghai 201306 , P.R. China
| | - Jing Yang
- c The Key Laboratory of Stem Cell Biology , Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) , Shanghai 200025 , P.R. China
| | - Tao Huang
- c The Key Laboratory of Stem Cell Biology , Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) , Shanghai 200025 , P.R. China
| | - Xiangyin Kong
- c The Key Laboratory of Stem Cell Biology , Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) and Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS) , Shanghai 200025 , P.R. China
| | - Lin Lu
- d Department of Radiology , Columbia University Medical Center , New York , NY 10032 , USA
| | - Yu-Dong Cai
- a College of Life Science , Shanghai University , Shanghai 200444 , P.R. China
| |
Collapse
|
191
|
Zhai L, Ma C, Li W, Yang S, Liu Z. miR-143 suppresses epithelial-mesenchymal transition and inhibits tumor growth of breast cancer through down-regulation of ERK5. Mol Carcinog 2015; 55:1990-2000. [PMID: 26618772 DOI: 10.1002/mc.22445] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 09/05/2015] [Accepted: 11/17/2015] [Indexed: 12/30/2022]
Abstract
Epithelial-mesenchymal transition (EMT) plays a pivotal role in the development of cancer invasion and metastasis. Many studies have significantly enhanced the knowledge on EMT through the characterization of microRNAs (miRNAs) influencing the signaling pathways and downstream events that define EMT on a molecular level. In this study, we found that miR-143 suppressed EMT. Up-regulating miR-143 enhanced E-cadherin-mediated cell-cell adhesion ability, reduced mesenchymal markers, and decreased cell proliferation, migration, and invasion in vitro. In vivo, the xenograft mouse model also unveiled the suppressive effects of miR-143 on tumor growth. Additionally, we demonstrated that up-regulating extracellular signal regulated kinase 5 (ERK5) was associated with poor prognosis of breast cancer patients. Moreover, we observed an inverse correlation between miR-143 and ERK5 in breast cancer tissues. miR-143 directly targeted seed sequences in the 3'-untranslated regions of ERK5. Furthermore, we revealed that the downstream molecules of glycogen synthase kinase 3 beta (GSK-3β)/Snail signaling were involved in EMT and modulated by ERK5. In summary, our findings demonstrated that miR-143 down-regulated its target ERK5, leading to the suppression of EMT induced by GSK-3β/Snail signaling of breast cancer. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Limin Zhai
- Department of Pathology, Weifang Medical University, Weifang, Shandong Province, P.R. China
| | - Chuanxiang Ma
- Department of Pathology, Afflidated Hospital of Weifang Medical University, Weifang, Shandong Province, P.R. China
| | - Wentong Li
- Department of Pathology, Weifang Medical University, Weifang, Shandong Province, P.R. China
| | - Shuo Yang
- Department of Pathology, Weifang Medical University, Weifang, Shandong Province, P.R. China
| | - Zhijun Liu
- Department of Medical Biology, Weifang Medical University, Weifang, Shandong Province, P.R. China
| |
Collapse
|
192
|
Cuadrado A. Structural and functional characterization of Nrf2 degradation by glycogen synthase kinase 3/β-TrCP. Free Radic Biol Med 2015; 88:147-157. [PMID: 25937177 DOI: 10.1016/j.freeradbiomed.2015.04.029] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/21/2015] [Accepted: 04/22/2015] [Indexed: 12/30/2022]
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a master regulator of cellular homeostasis that controls the expression of more than 1% of human genes related to biotransformation reactions, redox homeostasis, energetic metabolism, DNA repair, and proteostasis. Its activity has a tremendous impact on physiology and pathology and therefore it is very tightly regulated, mainly at the level of protein stability. In addition to the very well established regulation by the ubiquitin E3 ligase adapter Keap1, recent advances have identified a novel mechanism based on signaling pathways that regulate glycogen synthase kinse-3 (GSK-3). This kinase phosphorylates specific serine residues in the Neh6 domain of Nrf2 to create a degradation domain that is then recognized by the ubiquitin ligase adapter β-TrCP and tagged for proteasome degradation by a Cullin1/Rbx1 complex. Here we review the mechanistic elements and the signaling pathways that participate in this regulation by GSK-3/β-TrCP. These pathways include those activated by ligands of tyrosine kinase, G protein-coupled, metabotropic, and ionotropic receptors that activate phosphatidyl inositol 3-kinase (PI3K)/ATK and by the canonical WNT signaling pathway, where a fraction of Nrf2 interacts with Axin1/GSK-3. Considering that free Nrf2 protein is localized in the nucleus, we propose a model termed "double flux controller" to explain how Keap1 and β-TrCP coordinate the stability of Nrf2 in several scenarios. The GSK-3/β-TrCP axis provides a novel therapeutic strategy to modulate Nrf2 activity.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigaciones Biomedicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), and Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, 28029 Madrid, Spain.
| |
Collapse
|
193
|
So KY, Ahn SG, Oh SH. Autophagy regulated by prolyl isomerase Pin1 and phospho-Ser-GSK3αβ involved in protection of oral squamous cell carcinoma against cadmium toxicity. Biochem Biophys Res Commun 2015; 466:541-6. [DOI: 10.1016/j.bbrc.2015.09.066] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
|
194
|
Igea A, Nebreda AR. The Stress Kinase p38α as a Target for Cancer Therapy. Cancer Res 2015; 75:3997-4002. [PMID: 26377941 DOI: 10.1158/0008-5472.can-15-0173] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 05/19/2015] [Indexed: 12/11/2022]
Abstract
p38α is a ubiquitous protein kinase strongly activated by stress signals, inflammatory cytokines, and many other stimuli, which has been implicated in the modulation of multiple cellular processes. There is good evidence in the literature that p38α plays an important tumor-suppressor role by interfering with malignant cell transformation. This is mainly based on the ability of the p38α pathway to regulate tissue homeostasis by integrating signals that balance cell proliferation and differentiation or induce apoptosis. However, recent reports have also illustrated protumorigenic functions for p38α. Thus, p38α signaling may facilitate the survival and proliferation of tumor cells contributing to the progression of some tumor types. In addition, p38α activation helps tumor cells to survive chemotherapeutic treatments. In all these cases, the inhibition of p38α has a potential therapeutic interest. Further elucidation of the context-dependent functions of p38α signaling in tumoral processes is of obvious importance for the use of inhibitors of this pathway in cancer therapy.
Collapse
Affiliation(s)
- Ana Igea
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), Barcelona, Spain. Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
195
|
Zhang X, Yu X, Jiang G, Miao Y, Wang L, Zhang Y, Liu Y, Fan C, Lin X, Dong Q, Han Q, Zhao H, Han Y, Han X, Rong X, Ding S, Wang E, Wang E. Cytosolic TMEM88 Promotes Invasion and Metastasis in Lung Cancer Cells by Binding DVLS. Cancer Res 2015; 75:4527-37. [PMID: 26359454 DOI: 10.1158/0008-5472.can-14-3828] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 08/15/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Xiupeng Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xinmiao Yu
- Department of Surgical Oncology and Breast Surgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guiyang Jiang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yuan Miao
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Liang Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yong Zhang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yang Liu
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Chuifeng Fan
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xuyong Lin
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qianze Dong
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Qiang Han
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Huanyu Zhao
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yong Han
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xu Han
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xuezhu Rong
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Shuting Ding
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Endi Wang
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Enhua Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
196
|
Mier-Aguilar CA, Vega-Baray B, Burgueño-Bucio E, Lozano F, García-Zepeda EA, Raman C, Soldevila G. Functional requirement of tyrosine residue 429 within CD5 cytoplasmic domain for regulation of T cell activation and survival. Biochem Biophys Res Commun 2015; 466:381-7. [PMID: 26363459 DOI: 10.1016/j.bbrc.2015.09.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 09/05/2015] [Indexed: 11/15/2022]
Abstract
CD5 has been mainly described as a negative regulator of TCR and BCR signaling and recent evidence has shown an important role for this receptor in delivering pro-survival signals. However, the molecular mechanisms underlying these processes remain unresolved. TCR crosslinking leads to phosphorylation of three tyrosine residues within the cytoplasmic tail of CD5 (Y429, Y441 and Y463) leading to the recruitment of signaling molecules like PI3K, c-Cbl and RasGAP; nevertheless, the role of these residues in T cell survival has not yet been assessed. In this study, we show that alanine-scanning mutagenesis of such tyrosine residues, either singly or in combination, leads to an increased thymocyte cell death with or without α-CD3 stimulation. Remarkably, the T-cell death observed with each individual tyrosine mutant was Caspase 3-independent. Furthermore, Y429 mutation resulted in a hyper-phosphorylation of ERK suggesting that this tyrosine residue regulates cell survival through down modulation of TCR signaling. Mutation of Y441 or Y463 did not induce hyper-responsiveness to TCR activation, indicating that they promoted T-cell survival by a TCR signal-independent pathway. Our results show that three tyrosine-based domains within CD5 cytoplasmic tail promote T-cell survival through non-overlapping mechanisms. This study also reveals that Y429 domain of CD5, previously described as a "pseudo ITAM", is functionally an ITIM domain in T cells.
Collapse
Affiliation(s)
- Carlos A Mier-Aguilar
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, DF 04510, Mexico
| | - Benjamin Vega-Baray
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, DF 04510, Mexico
| | - Erica Burgueño-Bucio
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, DF 04510, Mexico
| | - Francisco Lozano
- Servei d'Immunologia, Hospital Clinic i Provincial de Barcelona, Institut d'Investigaciones Biomédiques August Pi i Sunyer (IDIBABS), Departament de Biologia Cel.lular, Immunologia i Neurociències, Universitat de Barcelona, Barcelona 08036, Spain
| | - Eduardo A García-Zepeda
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, DF 04510, Mexico
| | - Chander Raman
- Division of Clinical Immunology and Rheumatology, Departments of Medicine, and Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gloria Soldevila
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México, DF 04510, Mexico.
| |
Collapse
|
197
|
Liu W, Wang H, Wang Y, Li H, Ji L. Metabolic factors-triggered inflammatory response drives antidepressant effects of exercise in CUMS rats. Psychiatry Res 2015; 228:257-264. [PMID: 26144579 DOI: 10.1016/j.psychres.2015.05.102] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 05/15/2015] [Accepted: 05/25/2015] [Indexed: 12/22/2022]
Abstract
Chronic stress is a potential contributing factor for depression, accompanying with metabolic and inflammatory response. Exercise is considered as a treatment for depression, but mechanisms underlying its beneficial effects still remain unknown. The objectives of present study were to confirm that metabolic factors-triggered inflammatory response mediates the antidepressant actions of exercise in chronic unpredictable mild stress (CUMS) rats. It has been found that CUMS stimulated expression of ghrelin and its receptor Ghsr, but inhibited expression of leptin and its receptor LepRb. Ghrelin, via binding to Ghsr, induced phosphorylation of GSK-3β on Tyr216 and decreased phosphorylation on Ser9, thus increasing GSK-3β activity. Conversely, ghrelin binding to Ghsr decreased STAT3 activity, through decreasing phosphorylation of STAT3 on Tyr705 and increasing Ser727 phosphorylation. Negatively correlated with ghrelin, leptin binding to LepRb had opposite effects on the activity of GSK-3β and STAT3 via phosphorylation. In addition, decreased leptin level initiated NLRP3 activity via LepRb. Furthermore, GSK-3β inhibited STAT3 activation, thus promoting the expression of NLRP3. Meanwhile, swim improved metabolic and inflammatory response both in CUMS and control rats. Our findings suggest that exercise not only ameliorates metabolic disturbance and inflammatory response in depression, but also contributes to metabolic and inflammatory function in normal conditions.
Collapse
Affiliation(s)
- Weina Liu
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China.
| | - Hongmei Wang
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China
| | - Yangkai Wang
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Haipeng Li
- School of Physical Education & Health Care, Hangzhou Normal University, Hangzhou 311121, China
| | - Liu Ji
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China; School of Physical Education & Health Care, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
198
|
Schuettler D, Piontek G, Wirth M, Haller B, Reiter R, Brockhoff G, Pickhard A. Selective inhibition of EGFR downstream signaling reverses the irradiation-enhanced migration of HNSCC cells. Am J Cancer Res 2015; 5:2660-2672. [PMID: 26609474 PMCID: PMC4633896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 07/29/2015] [Indexed: 06/05/2023] Open
Abstract
Irradiation, which is one of the standard therapies used to treat squamous cell carcinoma of the head and neck (HNSCC), has been linked to enhanced tumor migration in carcinomas. In this study, we demonstrated that irradiation induced the phosphorylation of AKT, p38 MAPK and ERK. The combined activation of these pathways caused inactivation of GSK3β kinase, resulting in enhanced tumor cell migration. Here, we describe that the exclusive and specific inhibition of just one of the aforementioned key signaling molecules is sufficient to restore GSK3β activity and to reduce radiation-induced migration in HNSCC. These data indicate that pharmacological inhibition of pathways targeting GSK3β could decrease radiation-induced cell migration in HNSCC and thus potentially reduce metastasis and locoregional recurrence in patients.
Collapse
Affiliation(s)
- Dominik Schuettler
- Department of Otolaryngology Head and Neck Surgery, Technical University of MunichIsmaninger Str. 22, Muenchen 81675, Germany
| | - Guido Piontek
- Department of Otolaryngology Head and Neck Surgery, Technical University of MunichIsmaninger Str. 22, Muenchen 81675, Germany
| | - Markus Wirth
- Department of Otolaryngology Head and Neck Surgery, Technical University of MunichIsmaninger Str. 22, Muenchen 81675, Germany
| | - Bernhard Haller
- Institute for Medical Statistics and Epidemiology, Technical University of MunichIsmaninger Straße 22, Muenchen 81675, Germany
| | - Rudolf Reiter
- Department of Otolaryngology Head and Neck Surgery, Section of Phoniatrics and Pedaudiology, University of UlmPrittwitzstr. 43, Ulm 89075, Germany
| | - Gero Brockhoff
- Department of Gynecology and Obstetrics, University of RegensburgLandshuter Str. 65, Regensburg 93053, Germany
| | - Anja Pickhard
- Department of Otolaryngology Head and Neck Surgery, Technical University of MunichIsmaninger Str. 22, Muenchen 81675, Germany
| |
Collapse
|
199
|
NSC-87877 inhibits DUSP26 function in neuroblastoma resulting in p53-mediated apoptosis. Cell Death Dis 2015; 6:e1841. [PMID: 26247726 PMCID: PMC4558500 DOI: 10.1038/cddis.2015.207] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 06/22/2015] [Accepted: 06/30/2015] [Indexed: 01/07/2023]
Abstract
Dual specificity protein phosphatase 26 (DUSP26) is overexpressed in high-risk neuroblastoma (NB) and contributes to chemoresistance by inhibiting p53 function. In vitro, DUSP26 has also been shown to effectively inhibit p38 MAP kinase. We hypothesize that inhibiting DUSP26 will result in decreased NB cell growth in a p53 and/or p38-mediated manner. NSC-87877 (8-hydroxy-7-[(6-sulfo-2-naphthyl)azo]-5-quinolinesulfonic acid), a novel DUSP26 small molecule inhibitor, shows effective growth inhibition and induction of apoptosis in NB cell lines. NB cell lines treated with small hairpin RNA (shRNA) targeting DUSP26 also exhibit a proliferation defect both in vitro and in vivo. Treatment of NB cell lines with NSC-87877 results in increased p53 phosphorylation (Ser37 and Ser46) and activation, increased activation of downstream p38 effector proteins (heat shock protein 27 (HSP27) and MAP kinase-activated protein kinase 2 (MAPKAPK2)) and poly ADP ribose polymerase/caspase-3 cleavage. The cytotoxicity resulting from DUSP26 inhibition is partially reversed by knocking down p53 expression with shRNA and also by inhibiting p38 activity with SB203580 (4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine). In an intrarenal mouse model of NB, NSC-87877 treatment results in decreased tumor growth and increased p53 and p38 activity. Together, these results suggest that DUSP26 inhibition with NSC-87877 is an effective strategy to induce NB cell cytotoxicity in vitro and in vivo through activation of the p53 and p38 mitogen-activated protein kinase (MAPK) tumor-suppressor pathways.
Collapse
|
200
|
Resibufogenin Induces G1-Phase Arrest through the Proteasomal Degradation of Cyclin D1 in Human Malignant Tumor Cells. PLoS One 2015; 10:e0129851. [PMID: 26121043 PMCID: PMC4488249 DOI: 10.1371/journal.pone.0129851] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 05/13/2015] [Indexed: 11/19/2022] Open
Abstract
Huachansu, a traditional Chinese medicine prepared from the dried toad skin, has been used in clinical studies for various cancers in China. Resibufogenin is a component of huachansu and classified as bufadienolides. Resibufogenin has been shown to exhibit the anti-proliferative effect against cancer cells. However, the molecular mechanism of resibufogenin remains unknown. Here we report that resibufogenin induces G1-phase arrest with hypophosphorylation of retinoblastoma (RB) protein and down-regulation of cyclin D1 expression in human colon cancer HT-29 cells. Since the down-regulation of cyclin D1 was completely blocked by a proteasome inhibitor MG132, the suppression of cyclin D1 expression by resibufogenin was considered to be in a proteasome-dependent manner. It is known that glycogen synthase kinase-3β (GSK-3β) induces the proteasomal degradation of cyclin D1. The addition of GSK-3β inhibitor SB216763 inhibited the reduction of cyclin D1 caused by resibufogenin. These effects on cyclin D1 by resibufogenin were also observed in human lung cancer A549 cells. These findings suggest that the anti-proliferative effect of resibufogenin may be attributed to the degradation of cyclin D1 caused by the activation of GSK-3β.
Collapse
|