1
|
Domaniku-Waraich A, Agca S, Toledo B, Sucuoglu M, Özen SD, Bilgic SN, Arabaci DH, Kashgari AE, Kir S. Oncostatin M signaling drives cancer-associated skeletal muscle wasting. Cell Rep Med 2024; 5:101498. [PMID: 38569555 PMCID: PMC11031427 DOI: 10.1016/j.xcrm.2024.101498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 01/21/2024] [Accepted: 03/14/2024] [Indexed: 04/05/2024]
Abstract
Progressive weakness and muscle loss are associated with multiple chronic conditions, including muscular dystrophy and cancer. Cancer-associated cachexia, characterized by dramatic weight loss and fatigue, leads to reduced quality of life and poor survival. Inflammatory cytokines have been implicated in muscle atrophy; however, available anticytokine therapies failed to prevent muscle wasting in cancer patients. Here, we show that oncostatin M (OSM) is a potent inducer of muscle atrophy. OSM triggers cellular atrophy in primary myotubes using the JAK/STAT3 pathway. Identification of OSM targets by RNA sequencing reveals the induction of various muscle atrophy-related genes, including Atrogin1. OSM overexpression in mice causes muscle wasting, whereas muscle-specific deletion of the OSM receptor (OSMR) and the neutralization of circulating OSM preserves muscle mass and function in tumor-bearing mice. Our results indicate that activated OSM/OSMR signaling drives muscle atrophy, and the therapeutic targeting of this pathway may be useful in preventing muscle wasting.
Collapse
Affiliation(s)
| | - Samet Agca
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkiye
| | - Batu Toledo
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkiye
| | - Melis Sucuoglu
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkiye
| | - Sevgi Döndü Özen
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkiye
| | - Sevval Nur Bilgic
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkiye
| | - Dilsad Hilal Arabaci
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkiye
| | - Aynur Erkin Kashgari
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkiye
| | - Serkan Kir
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Turkiye.
| |
Collapse
|
2
|
Biltz RG, Swanson SP, Draime N, Davis AC, Yin W, Goodman EJ, Gallagher NR, Bhattacharya A, Sheridan JF, Godbout JP. Antagonism of the brain P2X7 ion channel attenuates repeated social defeat induced microglia reactivity, monocyte recruitment and anxiety-like behavior in male mice. Brain Behav Immun 2024; 115:356-373. [PMID: 37914101 PMCID: PMC10807695 DOI: 10.1016/j.bbi.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/18/2023] [Accepted: 10/14/2023] [Indexed: 11/03/2023] Open
Abstract
Chronic stress is linked to increased anxiety. Repeated social defeat (RSD) in mice causes anxiety that is dependent on activated neurons, reactive microglia, and accumulation of monocytes in the brain. This response requires interactions between the immune system and central nervous system (CNS). Neuronal activation within threat appraisal regions is a key response to RSD, however, it is unclear how microglia become activated. One potential explanation is that microglia express a purinergic non-selective ligand gated adenosine-triphosphate (ATP) receptor 7 (P2X7). Activation of P2X7 promotes the release of chemokines and cytokines, and recruitment of monocytes to the brain. Thus, the purpose of this study was to determine if a novel P2X7 antagonist blocked neuronal and microglia interactions and the corresponding anxiety following RSD. Male mice were administered (i.p.) a P2X7 antagonist, JNJ-54471300, prior to each cycle of RSD. Fourteen hours after RSD, behavioral deficits including social avoidance and anxiety-like were determined. Moreover, several immune parameters were assessed. RSD caused neuronal activation in stress-responsive regions, monocyte production and release, splenomegaly, and social avoidance. These parameters were unaffected by P2X7 antagonism. RSD-associated proportional area of Iba-1+ microglia, monocyte accumulation in the brain, IL-1β mRNA expression in enriched myeloid cells, plasma IL-6, and anxiety-like behavior were ameliorated by P2X7 antagonism. Gene expression analysis in the hippocampus and amygdala showed regional specific responses to RSD and some were reversed with P2X7 antagonism. Overall, blocking P2X7 activation attenuated RSD-induced microglia reactivity with corresponding reduction in neuroinflammation, monocyte accumulation, and anxiety-like behavior in male mice.
Collapse
Affiliation(s)
- Rebecca G Biltz
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, United States
| | - Samuel P Swanson
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, United States
| | - Natalie Draime
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, United States
| | - Amara C Davis
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, United States
| | - Wenyuan Yin
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, United States
| | - Ethan J Goodman
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, United States
| | - Natalie R Gallagher
- Division of Biosciences, The Ohio State University College of Dentistry, United States; Institute for Behavioral Medicine Research, The Ohio State University, Wexner Medical Center, United States
| | - Anindya Bhattacharya
- Neuroscience, Janssen Research and Development, LLC, San Diego, CA, United States
| | - John F Sheridan
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, United States; Division of Biosciences, The Ohio State University College of Dentistry, United States; Chronic Brain Injury Program, The Ohio State University, United States; Institute for Behavioral Medicine Research, The Ohio State University, Wexner Medical Center, United States.
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University, Wexner Medical Center, United States; Chronic Brain Injury Program, The Ohio State University, United States; Institute for Behavioral Medicine Research, The Ohio State University, Wexner Medical Center, United States.
| |
Collapse
|
3
|
Díaz-Chamorro S, Garrido-Jiménez S, Barrera-López JF, Mateos-Quirós CM, Cumplido-Laso G, Lorenzo MJ, Román ÁC, Bernardo E, Sabio G, Carvajal-González JM, Centeno F. Title: p38δ Regulates IL6 Expression Modulating ERK Phosphorylation in Preadipocytes. Front Cell Dev Biol 2022; 9:708844. [PMID: 35111744 PMCID: PMC8802314 DOI: 10.3389/fcell.2021.708844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 12/10/2021] [Indexed: 11/13/2022] Open
Abstract
IL6 is an essential cytokine in metabolism regulation and for intercommunication among different organs and tissues. IL6 produced by different tissues has different functions and therefore it is very important to understand the mechanism of its expression in adipose tissue. In this work we demonstrated that IL6 expression in mouse preadipocytes, like in human, is partially dependent on Wnt5a and JNK. Using mouse preadipocytes lacking each one of the p38 SAPK family members, we have shown that IL6 expression is also p38γ and p38δ dependent. In fact, the lack of some of these two kinases increases IL6 expression without altering that of Wnt5a. Moreover, we show that the absence of p38δ promotes greater ERK1/2 phosphorylation in a MEK1/2 independent manner, and that this increased ERK1/2 phosphorylation state is contributing to the higher IL6 expression in p38δ−/- preadipocytes. These results suggest a new crosstalk between two MAPK signaling pathway, p38δ and ERK1/2, where p38δ modulates the phosphorylation state of ERK1/2.
Collapse
Affiliation(s)
- Selene Díaz-Chamorro
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Sergio Garrido-Jiménez
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Juan Francisco Barrera-López
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Clara María Mateos-Quirós
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Guadalupe Cumplido-Laso
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - María Jesús Lorenzo
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Cáceres, Spain
| | - Ángel Carlos Román
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Edgar Bernardo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - José María Carvajal-González
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| | - Francisco Centeno
- Universidad de Extremadura, Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Ciencias, Badajoz, Spain
| |
Collapse
|
4
|
Saad OA, Li WT, Krishnan AR, Nguyen GC, Lopez JP, McKay RR, Wang-Rodriguez J, Ongkeko WM. The renal clear cell carcinoma immune landscape. Neoplasia 2022; 24:145-154. [PMID: 34991061 PMCID: PMC8740459 DOI: 10.1016/j.neo.2021.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/20/2021] [Indexed: 01/05/2023]
Abstract
A comprehensive evaluation of the clear cell renal cell carcinoma (ccRCC) immune landscape was found using 584 RNA-sequencing datasets from The Cancer Genome Atlas (TCGA), we identified 17 key dysregulated immune-associated genes in ccRCC based on association with clinical variables and important immune pathways. Of the numerous findings from our analyses, we found that several of the 17 key dysregulated genes are heavily involved in interleukin and NF-kB signaling and that somatic copy number alteration (SCNA) hotspots may be causally associated with gene dysregulation. More importantly, we also found that key immune-associated genes and pathways are strongly upregulated in ccRCC. Our study may lend novel insights into the clinical implications of immune dysregulation in ccRCC and suggests potential immunotherapeutic targets for further evaluation.
Collapse
Affiliation(s)
- Omar A Saad
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Wei Tse Li
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Aswini R Krishnan
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Griffith C Nguyen
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Jay P Lopez
- Division of Hematology and Oncology, New York-Presbyterian/Weill Cornell Medical College, NY, USA
| | - Rana R McKay
- Department of Medicine, Division of Hematology/Oncology, University of California San Diego, San Diego, CA, USA.
| | - Jessica Wang-Rodriguez
- Veterans Administration Medical Center and Department of Pathology, University of California, San Diego, La Jolla, California 92161, USA.
| | - Weg M Ongkeko
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
5
|
Fraszczak J, Arman KM, Lacroix M, Vadnais C, Gaboury L, Möröy T. Severe Inflammatory Reactions in Mice Expressing a GFI1 P2A Mutant Defective in Binding to the Histone Demethylase KDM1A (LSD1). THE JOURNAL OF IMMUNOLOGY 2021; 207:1599-1615. [PMID: 34408010 DOI: 10.4049/jimmunol.2001146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 07/07/2021] [Indexed: 11/19/2022]
Abstract
GFI1 is a DNA-binding transcription factor that regulates hematopoiesis by repressing target genes through its association with complexes containing histone demethylases such as KDM1A (LSD1) and histone deacetylases (HDACs). To study the consequences of the disruption of the complex between GFI1 and histone-modifying enzymes, we have used knock-in mice harboring a P2A mutation in GFI1 coding region that renders it unable to bind LSD1 and associated histone-modifying enzymes such as HDACs. GFI1P2A mice die prematurely and show increased numbers of memory effector and regulatory T cells in the spleen accompanied by a severe systemic inflammation with high serum levels of IL-6, TNF-α, and IL-1β and overexpression of the gene encoding the cytokine oncostatin M (OSM). We identified lung alveolar macrophages, CD8 T cell from the spleen and thymic eosinophils, and monocytes as the sources of these cytokines in GFI1P2A mice. Chromatin immunoprecipitation showed that GFI1/LSD1 complexes occupy sites at the Osm promoter and an intragenic region of the Tnfα gene and that a GFI1P2A mutant still remains bound at these sites even without LSD1. Methylation and acetylation of histone H3 at these sites were enriched in cells from GFI1P2A mice, the H3K27 acetylation being the most significant. These data suggest that the histone modification facilitated by GFI1 is critical to control inflammatory pathways in different cell types, including monocytes and eosinophils, and that a disruption of GFI1-associated complexes can lead to systemic inflammation with fatal consequences.
Collapse
Affiliation(s)
| | - Kaifee Mohammad Arman
- Institut de Recherches Cliniques de Montréal, Montreal, Canada.,Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Marion Lacroix
- Institut de Recherches Cliniques de Montréal, Montreal, Canada.,Division of Experimental Medicine, McGill University, Montreal, Canada
| | - Charles Vadnais
- Institut de Recherches Cliniques de Montréal, Montreal, Canada
| | - Louis Gaboury
- Unité de Recherche en Histologie et Pathologie Moléculaire, Institut de Recherche en Immunologie et en Cancérologie, Montreal, Canada.,Département de Pathologie et Biologie Cellulaire, Faculté de Médecine, Université de Montréal, Montreal, Canada; and
| | - Tarik Möröy
- Institut de Recherches Cliniques de Montréal, Montreal, Canada; .,Division of Experimental Medicine, McGill University, Montreal, Canada.,Département de Microbiologie Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montreal, Canada
| |
Collapse
|
6
|
Neuroprotective Effects of Milrinone on Experimental Acute Spinal Cord Injury: Rat Model. World Neurosurg 2021; 147:e225-e233. [DOI: 10.1016/j.wneu.2020.12.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 10/22/2022]
|
7
|
Sharanek A, Burban A, Laaper M, Heckel E, Joyal JS, Soleimani VD, Jahani-Asl A. OSMR controls glioma stem cell respiration and confers resistance of glioblastoma to ionizing radiation. Nat Commun 2020; 11:4116. [PMID: 32807793 PMCID: PMC7431428 DOI: 10.1038/s41467-020-17885-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 07/22/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma contains a rare population of self-renewing brain tumor stem cells (BTSCs) which are endowed with properties to proliferate, spur the growth of new tumors, and at the same time, evade ionizing radiation (IR) and chemotherapy. However, the drivers of BTSC resistance to therapy remain unknown. The cytokine receptor for oncostatin M (OSMR) regulates BTSC proliferation and glioblastoma tumorigenesis. Here, we report our discovery of a mitochondrial OSMR that confers resistance to IR via regulation of oxidative phosphorylation, independent of its role in cell proliferation. Mechanistically, OSMR is targeted to the mitochondrial matrix via the presequence translocase-associated motor complex components, mtHSP70 and TIM44. OSMR interacts with NADH ubiquinone oxidoreductase 1/2 (NDUFS1/2) of complex I and promotes mitochondrial respiration. Deletion of OSMR impairs spare respiratory capacity, increases reactive oxygen species, and sensitizes BTSCs to IR-induced cell death. Importantly, suppression of OSMR improves glioblastoma response to IR and prolongs lifespan. The suppression of the receptor for oncostatin M (OSMR) can prevent glioblastoma cell growth. Here, the authors demonstrate a role for OSMR in modulating glioma stem cell respiration and its impact on resistance to ionizing radiation.
Collapse
Affiliation(s)
- Ahmad Sharanek
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Audrey Burban
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada
| | - Matthew Laaper
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada.,Integrated program in Neuroscience, Montreal Neurological Institute, 3801 University Street, Montréal, QC, H3A 2B4, Canada
| | - Emilie Heckel
- Departments of Pediatrics, Pharmacology and Ophthalmology, Université de Montréal, CHU Sainte-Justine, Montréal, QC, H3T 1C5, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Jean-Sebastien Joyal
- Departments of Pediatrics, Pharmacology and Ophthalmology, Université de Montréal, CHU Sainte-Justine, Montréal, QC, H3T 1C5, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Vahab D Soleimani
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada.,Department of Human Genetics, McGill University, 3640 Rue University, Montréal, QC, H3A OC7, Canada
| | - Arezu Jahani-Asl
- Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1E2, Canada. .,Integrated program in Neuroscience, Montreal Neurological Institute, 3801 University Street, Montréal, QC, H3A 2B4, Canada. .,Gerald Bronfman Department of Oncology and Division of Experimental Medicine, McGill University, 5100 Maisonneuve Blvd West, Suite 720, H4A3T2, Montréal, QC, Canada.
| |
Collapse
|
8
|
Cash A, Theus MH. Mechanisms of Blood-Brain Barrier Dysfunction in Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21093344. [PMID: 32397302 PMCID: PMC7246537 DOI: 10.3390/ijms21093344] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injuries (TBIs) account for the majority of injury-related deaths in the United States with roughly two million TBIs occurring annually. Due to the spectrum of severity and heterogeneity in TBIs, investigation into the secondary injury is necessary in order to formulate an effective treatment. A mechanical consequence of trauma involves dysregulation of the blood–brain barrier (BBB) which contributes to secondary injury and exposure of peripheral components to the brain parenchyma. Recent studies have shed light on the mechanisms of BBB breakdown in TBI including novel intracellular signaling and cell–cell interactions within the BBB niche. The current review provides an overview of the BBB, novel detection methods for disruption, and the cellular and molecular mechanisms implicated in regulating its stability following TBI.
Collapse
Affiliation(s)
- Alison Cash
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA;
| | - Michelle H. Theus
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA;
- The Center for Regenerative Medicine, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
- Correspondence: ; Tel.: 1-540-231-0909; Fax: 1-540-231-7425
| |
Collapse
|
9
|
Mohammadi C, Sameri S, Najafi R. Insight into adipokines to optimize therapeutic effects of stem cell for tissue regeneration. Cytokine 2020; 128:155003. [PMID: 32000014 DOI: 10.1016/j.cyto.2020.155003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 11/29/2022]
Abstract
Stem cell therapy is considered as a promising regenerative medicine for repairing and treating damaged tissues and/or preventing various diseases. But there are still some obstacles such as low cell migration, poor stem cell engraftment and decreased cell survival that need to be overcome before transplantation. Therefore, a large body of studies has focused on improving the efficiency of stem cell therapy. For instance, preconditioning of stem cells has emerged as an effective strategy to reinforce therapeutic efficacy. Adipokines are signaling molecules, secreted by adipose tissue, which regulate a variety of biological processes in adipose tissue and other organs including the brain, liver, and muscle. In this review article, we shed light on the biological effects of some adipokines including apelin, oncostatin M, omentin-1 and vaspin on stem cell therapy and the most recent preclinical advances in our understanding of how these functions ameliorate stem cell therapy outcome.
Collapse
Affiliation(s)
- Chiman Mohammadi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Saba Sameri
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
10
|
Prokaryotic soluble overexpression and purification of oncostatin M using a fusion approach and genetically engineered E. coli strains. Sci Rep 2019; 9:13706. [PMID: 31548569 PMCID: PMC6757106 DOI: 10.1038/s41598-019-50110-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 09/06/2019] [Indexed: 12/16/2022] Open
Abstract
Human Oncostatin M (OSM), initially discovered as a tumour inhibitory factor secreted from U-937 cells, is a gp130 (IL-6/LIF) cytokine family member that exhibits pleiotropic effects in inflammation, haematopoiesis, skeletal tissue alteration, liver regeneration, cardiovascular and metabolic diseases. Cytoplasmic expression of OSM in Escherichia coli results in inclusion bodies, and complex solubilisation, refolding and purification is required to prepare bioactive protein. Herein, eight N-terminal fusion variants of OSM with hexahistidine (His6) tag and seven solubility-enhancing tags, including thioredoxin (Trx), small ubiquitin-related modifier (Sumo), glutathione S-transferase (GST), maltose-binding protein (MBP), N-utilisation substance protein A (Nusa), human protein disulphide isomerase (PDI) and the b‘a’ domain of PDI (PDIb‘a’), were tested for soluble OSM expression in E. coli. The His6-OSM plasmid was also introduced into genetically engineered Origami 2 and SHuffle strains to test expression of the protein. At 18 °C, MBP-tagged OSM was highly expressed and solubility was dramatically enhanced. In addition, His6-OSM was more highly expressed and soluble in Origami 2 and SHuffle strains than in BL21(DE3). MBP-OSM and His6-OSM were purified more than 95% with yields of 11.02 mg and 3.27 mg from a 500 mL culture. Protein identity was confirmed by mass spectroscopy, and bioactivity was demonstrated by in vitro inhibition of Th17 cell differentiation.
Collapse
|
11
|
Houben E, Hellings N, Broux B. Oncostatin M, an Underestimated Player in the Central Nervous System. Front Immunol 2019; 10:1165. [PMID: 31191538 PMCID: PMC6549448 DOI: 10.3389/fimmu.2019.01165] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022] Open
Abstract
For a long time, the central nervous system (CNS) was believed to be an immune privileged organ. In the last decades, it became apparent that the immune system interacts with the CNS not only in pathological, but also in homeostatic situations. It is now clear that immune cells infiltrate the healthy CNS as part of immune surveillance and that immune cells communicate through cytokines with CNS resident cells. In pathological conditions, an enhanced infiltration of immune cells takes place to fight the pathogen. A well-known family of cytokines is the interleukin (IL)-6 cytokine family. All members are important in cell communication and cell signaling in the immune system. One of these members is oncostatin M (OSM), for which the receptor is expressed on several cells of the CNS. However, the biological function of OSM in the CNS is not studied in detail. Here, we briefly describe the general aspects related to OSM biology, including signaling and receptor binding. Thereafter, the current understanding of OSM during CNS homeostasis and pathology is summarized.
Collapse
Affiliation(s)
- Evelien Houben
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Niels Hellings
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Bieke Broux
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
12
|
Tawara K, Scott H, Emathinger J, Wolf C, LaJoie D, Hedeen D, Bond L, Montgomery P, Jorcyk C. HIGH expression of OSM and IL-6 are associated with decreased breast cancer survival: synergistic induction of IL-6 secretion by OSM and IL-1β. Oncotarget 2019; 10:2068-2085. [PMID: 31007849 PMCID: PMC6459341 DOI: 10.18632/oncotarget.26699] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 01/31/2019] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation has been recognized as a risk factor for the development and maintenance of malignant disease. Cytokines such as interleukin-6 (IL-6), oncostatin M (OSM), and interleukin-1 beta (IL-1β) promote the development of both acute and chronic inflammation while promoting in vitro metrics of breast cancer metastasis. However, anti-IL-6 and anti-IL-1β therapeutics have not yielded significant results against solid tumors in clinical trials. Here we show that these three cytokines are interrelated in expression. Using the Curtis TCGA™ dataset, we have determined that there is a correlation between expression levels of OSM, IL-6, and IL-1β and reduced breast cancer patient survival (r = 0.6, p = 2.2 x 10−23). Importantly, we confirm that OSM induces at least a 4-fold increase in IL-6 production from estrogen receptor-negative (ER−) breast cancer cells in a manner that is dependent on STAT3 signaling. Furthermore, OSM induces STAT3 phosphorylation and IL-1β promotes p65 phosphorylation to synergistically induce IL-6 secretion in ER− MDA-MB-231 and to a lesser extent in ER+ MCF7 human breast cancer cells. Induction may be reduced in the ER+ MCF7 cells due to a previously known suppressive interaction between ER and STAT3. Interestingly, we show in MCF7 cells that ER’s interaction with STAT3 is reduced by 50% through both OSM and IL-1β treatment, suggesting a role for ER in mitigating STAT3-mediated inflammatory cascades. Here, we provide a rationale for a breast cancer treatment regime that simultaneously suppresses multiple targets, as these cytokines possess many overlapping functions that increase metastasis and worsen patient survival.
Collapse
Affiliation(s)
- Ken Tawara
- Boise State University, Biomolecular Sciences Program, Boise, ID, USA
| | - Hannah Scott
- Boise State University, Department of Biological Sciences, Boise, ID, USA
| | | | - Cody Wolf
- Boise State University, Biomolecular Sciences Program, Boise, ID, USA.,Boise State University, Department of Biological Sciences, Boise, ID, USA
| | - Dollie LaJoie
- Boise State University, Department of Biological Sciences, Boise, ID, USA.,University of Utah, Department of Oncological Sciences, Salt Lake City, UT, USA
| | - Danielle Hedeen
- Boise State University, Department of Biological Sciences, Boise, ID, USA.,University of Utah, Department of Oncological Sciences, Salt Lake City, UT, USA
| | - Laura Bond
- Boise State University, Biomolecular Research Center, Boise, ID, USA
| | | | - Cheryl Jorcyk
- Boise State University, Biomolecular Sciences Program, Boise, ID, USA.,Boise State University, Department of Biological Sciences, Boise, ID, USA
| |
Collapse
|
13
|
Han J, Feng Z, Xie Y, Li F, Lv B, Hua T, Zhang Z, Sun C, Su D, Ouyang Q, Cai Y, Zou Y, Tang Y, Sun H, Jiang X. Oncostatin M-induced upregulation of SDF-1 improves Bone marrow stromal cell migration in a rat middle cerebral artery occlusion stroke model. Exp Neurol 2019; 313:49-59. [DOI: 10.1016/j.expneurol.2018.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 09/03/2018] [Accepted: 09/07/2018] [Indexed: 01/02/2023]
|
14
|
An ALPHA7 Nicotinic Acetylcholine Receptor Agonist (GTS-21) Promotes C2C12 Myonuclear Accretion in Association with Release of Interleukin-6 (IL-6) and Improves Survival in Burned Mice. Shock 2018; 48:227-235. [PMID: 28282360 DOI: 10.1097/shk.0000000000000849] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The role of interleukin-6 (IL-6) in physiological processes and disease is poorly understood. The hypothesis tested in this study was that selective alpha7 acetylcholine receptor (α7AChR) agonist, GTS-21, releases IL-6 in association with myonuclear accretion and enhances insulin signaling in muscle cells, and improves survival of burn injured (BI) mice. The in vitro effects of GTS-21 were determined in C2C12 myoblasts and 7-day differentiated myotubes (myotubes). The in vivo effects of GTS-21 were tested in BI wild-type (WT) and IL-6 knockout (IL6KO) mice. GTS-21 dose-dependently (0 μM, 100 μM, and 200 μM) significantly increased IL-6 levels in myoblasts and myotubes at 6 and 9 h. GTS-21-induced IL-6 release in myotubes was attenuated by methyllycaconitine (α7AChR antagonist), and by Stat-3 or Stat-5 inhibitors. GTS-21 increased MyoD and Pax7 protein expression, myonuclear accretion, and insulin-induced phosphorylation of Akt, GSK-3β, and Glut4 in myotubes. The glucose levels of burned IL6KO mice receiving GTS-21 decreased significantly compared with sham-burn IL6KO mice. Superimposition of BI on IL6KO mice caused 100% mortality; GTS-21 reduced mortality to 75% in the IL6KO mice. The 75% mortality in burned WT mice was reduced to 0% with GTS-21. The in vitro findings suggest that GTS-21-induced IL-6 release from muscle is mediated via α7AChRs upstream of Stat-3 and -5 pathways and is associated with myonuclear accretion, possibly via MyoD and Pax7 expression. GTS-21 in vivo improves survival in burned WT mice and IL6KO mice, suggesting a potential therapeutic application of α7AChR agonists in the treatment of BI.
Collapse
|
15
|
Chuerduangphui J, Ekalaksananan T, Chaiyarit P, Patarapadungkit N, Chotiyano A, Kongyingyoes B, Promthet S, Pientong C. Effects of arecoline on proliferation of oral squamous cell carcinoma cells by dysregulating c-Myc and miR-22, directly targeting oncostatin M. PLoS One 2018; 13:e0192009. [PMID: 29385191 PMCID: PMC5791990 DOI: 10.1371/journal.pone.0192009] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/15/2018] [Indexed: 12/19/2022] Open
Abstract
Arecoline, the major alkaloid of areca nut, is known to induce oral carcinogenesis, however, its mechanism is still needed to elucidate. This study investigated the effects of arecoline on cell viability and cell-cycle progression of oral squamous cell carcinoma (OSCC) cells as well as a relevant cellular gene expression. The results showed that a low concentration of arecoline (0.025 μg/ml) increased OSCC cell viability, proportion of cells in G2/M phase and cell proliferation. Simultaneously, it induced IL-6, STAT3 and c-Myc expression. Interestingly, c-myc promoter activity was also induced by arecoline. MiR-22 expression in arecoline-treated OSCC cells was suppressed and comparable to an upregulated c-Myc expression. In arecoline-treated OSCC cells, oncostatin M (OSM) expression was significantly upregulated and inversely correlated with miR-22 expression. Likewise, OSM expression and its post-transcriptional activity were significantly decreased in miR-22-transfected OSCC and 293FT cells. This result demonstrated that miR-22 directly targeted OSM. Interestingly, miR-22 played an important role as a tumor suppresser on suppressing cell proliferation, migration and cell-cycle progression of OSCC cells. This result suggested the effect of arecoline to promote cell proliferation and cell-cycle progression of OSCC cells might be involved in induction of c-Myc expression and reduction of miR-22 resulting in OSM upregulation.
Collapse
Affiliation(s)
- Jureeporn Chuerduangphui
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Ponlatham Chaiyarit
- Department of Oral Diagnosis, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand.,Research Group of Chronic Inflammatory Oral Diseases and Systemic Diseases Associated with Oral Health, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Natcha Patarapadungkit
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand.,Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Chotiyano
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand.,Anatomical Pathology Unit, Khon Kaen Hospital, Khon Kaen, Thailand
| | - Bunkerd Kongyingyoes
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Supannee Promthet
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand.,Department of Epidemiology, Faculty of Public Health, Khon Kaen University, Khon Kaen, Thailand.,ASEAN Cancer Epidemiology and Prevention Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
16
|
Leifheit-Nestler M, Haffner D. Paracrine Effects of FGF23 on the Heart. Front Endocrinol (Lausanne) 2018; 9:278. [PMID: 29892269 PMCID: PMC5985311 DOI: 10.3389/fendo.2018.00278] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/11/2018] [Indexed: 12/17/2022] Open
Abstract
Fibroblast growth factor (FGF) 23 is a phosphaturic hormone primarily secreted by osteocytes to maintain phosphate and mineral homeostasis. In patients with and without chronic kidney disease, enhanced circulating FGF23 levels associate with pathologic cardiac remodeling, i.e., left ventricular hypertrophy (LVH) and myocardial fibrosis and increased cardiovascular mortality. Experimental studies demonstrate that FGF23 promotes hypertrophic growth of cardiac myocytes via FGF receptor 4-dependent activation of phospholipase Cγ/calcineurin/nuclear factor of activated T cell signaling independent of its co-receptor klotho. Recent studies indicate that FGF23 is also expressed in the heart, and markedly enhanced in various clinical and experimental settings of cardiac remodeling and heart failure independent of preserved or reduced renal function. On a cellular level, FGF23 is expressed in cardiac myocytes and in other non-cardiac myocytes, including cardiac fibroblasts, vascular smooth muscle and endothelial cells in coronary arteries, and in inflammatory macrophages. Current data suggest that secreted by cardiac myocytes, FGF23 can stimulate pro-fibrotic factors in myocytes to induce fibrosis-related pathways in fibroblasts and consequently cardiac fibrosis in a paracrine manner. While acting on cardiac myocytes, FGF23 directly induces pro-hypertrophic genes and promotes the progression of LVH in an autocrine and paracrine fashion. Thus, enhanced FGF23 may promote cardiac injury in various clinical settings not only by endocrine but also via paracrine/autocrine mechanisms. In this review, we discuss recent clinical and experimental data regarding molecular mechanisms of FGF23's paracrine action on the heart with respect to pathological cardiac remodeling.
Collapse
|
17
|
Ito K, Noguchi A, Uosaki Y, Taga T, Arakawa H, Takizawa T. Gfap and Osmr regulation by BRG1 and STAT3 via interchromosomal gene clustering in astrocytes. Mol Biol Cell 2017; 29:209-219. [PMID: 29142070 PMCID: PMC5909932 DOI: 10.1091/mbc.e17-05-0271] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 10/11/2017] [Accepted: 11/08/2017] [Indexed: 01/09/2023] Open
Abstract
Gene clustering is relevant in the regulation of gene expression. However, the mechanisms of gene clustering remain to be elucidated. Using a glial differentiation system, we found that the clustering of Gfap, an astrocyte-pecific gene, with Osmr enhances transcription of both genes. BRG1 and the JAK-STAT pathway are central to the clustering. Long-range chromatin interactions between gene loci in the cell nucleus are important for many biological processes, including transcriptional regulation. Previously, we demonstrated that several genes specifically cluster with the astrocyte-specific gene for glial fibrillary acidic protein (Gfap) during astrocyte differentiation; however, the molecular mechanisms for gene clustering remain largely unknown. Here we show that brahma-related gene 1 (BRG1), an ATP-dependent chromatin remodeling factor, and the transcription factor STAT3 are required for Gfap and oncostatin M receptor (Osmr) clustering and enhanced expression through recruitment to STAT3 recognition sequences and that gene clustering occurs prior to transcriptional up-regulation. BRG1 knockdown and JAK-STAT signaling inhibition impaired clustering, leading to transcriptional down-regulation of both genes. BRG1 and STAT3 were recruited to the same Gfap fragment; JAK-STAT signaling inhibition impaired BRG1 recruitment. Our results suggest that BRG1 and STAT3 coordinately regulate gene clustering and up-regulate Gfap and Osmr transcription.
Collapse
Affiliation(s)
- Kenji Ito
- Department of Pediatrics, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan.,Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Sakyoku, Kyoto 606 8507, Japan
| | - Azumi Noguchi
- Department of Pediatrics, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan
| | - Yuichi Uosaki
- Department of Pediatrics, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan
| | - Testuya Taga
- Department of Stem Cell Regulation, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-Ku, Tokyo 113-8510, Japan
| | - Hirokazu Arakawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan
| | - Takumi Takizawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan
| |
Collapse
|
18
|
Riemann A, Reime S, Thews O. Tumor Acidosis and Hypoxia Differently Modulate the Inflammatory Program: Measurements In Vitro and In Vivo. Neoplasia 2017; 19:1033-1042. [PMID: 29149667 PMCID: PMC5695649 DOI: 10.1016/j.neo.2017.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/29/2017] [Accepted: 09/29/2017] [Indexed: 12/29/2022] Open
Abstract
Inflammatory mediators produced by the tumor cells are of importance for immune response but also for malignant progression. The aim of the study was to analyze the expression of monocyte chemoattractant protein-1, interleukin-6 (IL-6), tumor necrosis factor-α, inducible isoform of nitric oxide synthase (iNOS), cyclooxygenase-2, and osteopontin in vitro in two different tumor cell lines under hypoxia (pO2 ≈ 1.5 mmHg) and/or acidosis (pH = 6.6) for up to 24 hours since hypoxia and acidosis are common characteristics of solid tumors. Additionally, the same tumor cell lines implanted in vivo were made hypoxic and acidotic artificially for 24 hours, after which the cytokine expression was measured. Finally, the activation of ERK1/2 and p38 by acidosis/hypoxia and their impact on cytokine expression were studied. The results indicate that acidosis and hypoxia have fundamentally different (often opposing) effects on cytokine expression. In addition, these effects were tumor cell line specific. When combining hypoxia and acidosis, the overall changes reflect an additive effect of both conditions alone, indicating that hypoxia and acidosis act by independent mechanisms. The in vivo changes corresponded well with the results obtained in the isolated tumor cells. Only iNOS expression was downregulated in vivo but increased in cell culture. For IL-6 expression, the acidosis-induced changes were dependent on ERK1/2 activation. In conclusion, it was demonstrated that the environmental pO2 and pH strongly affect the expression of inflammatory mediators in tumor cells. In vivo, most of the inflammatory mediators were downregulated, which could limit the activation of immune cells and by this foster the immune escape of tumors.
Collapse
Affiliation(s)
- Anne Riemann
- Julius Bernstein Institute of Physiology, University Halle-Wittenberg, Germany.
| | - Sarah Reime
- Julius Bernstein Institute of Physiology, University Halle-Wittenberg, Germany
| | - Oliver Thews
- Julius Bernstein Institute of Physiology, University Halle-Wittenberg, Germany
| |
Collapse
|
19
|
Magistri M, Khoury N, Mazza EMC, Velmeshev D, Lee JK, Bicciato S, Tsoulfas P, Faghihi MA. A comparative transcriptomic analysis of astrocytes differentiation from human neural progenitor cells. Eur J Neurosci 2016; 44:2858-2870. [PMID: 27564458 DOI: 10.1111/ejn.13382] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/25/2016] [Accepted: 08/23/2016] [Indexed: 12/11/2022]
Abstract
Astrocytes are a morphologically and functionally heterogeneous population of cells that play critical roles in neurodevelopment and in the regulation of central nervous system homeostasis. Studies of human astrocytes have been hampered by the lack of specific molecular markers and by the difficulties associated with purifying and culturing astrocytes from adult human brains. Human neural progenitor cells (NPCs) with self-renewal and multipotent properties represent an appealing model system to gain insight into the developmental genetics and function of human astrocytes, but a comprehensive molecular characterization that confirms the validity of this cellular system is still missing. Here we used an unbiased transcriptomic analysis to characterize in vitro culture of human NPCs and to define the gene expression programs activated during the differentiation of these cells into astrocytes using FBS or the combination of CNTF and BMP4. Our results demonstrate that in vitro cultures of human NPCs isolated during the gliogenic phase of neurodevelopment mainly consist of radial glial cells (RGCs) and glia-restricted progenitor cells. In these cells the combination of CNTF and BMP4 activates the JAK/STAT and SMAD signaling cascades, leading to the inhibition of oligodendrocytes lineage commitment and activation of astrocytes differentiation. On the other hand, FBS-derived astrocytes have properties of reactive astrocytes. Our work suggests that in vitro culture of human NPCs represents a valuable cellular system to study human disorders characterized by impairment of astrocytes development and function. Our datasets represent an important resource for researchers studying human astrocytes development and might set the basis for the discovery of novel human-specific astrocyte markers.
Collapse
Affiliation(s)
- Marco Magistri
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| | - Nathalie Khoury
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| | - Emilia Maria Cristina Mazza
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Dmitry Velmeshev
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvio Bicciato
- Department of Life Sciences, Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Pantelis Tsoulfas
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mohammad Ali Faghihi
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, University of Miami Miller School of Medicine, 1501 NW 10th Ave, BRB 508, Miami, FL, 33136, USA
| |
Collapse
|
20
|
Cytokine and Growth Factor Activation In Vivo and In Vitro after Spinal Cord Injury. Mediators Inflamm 2016; 2016:9476020. [PMID: 27418745 PMCID: PMC4935915 DOI: 10.1155/2016/9476020] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/18/2016] [Indexed: 12/22/2022] Open
Abstract
Spinal cord injury results in a life-disrupting series of deleterious interconnected mechanisms encompassed by the primary and secondary injury. These events are mediated by the upregulation of genes with roles in inflammation, transcription, and signaling proteins. In particular, cytokines and growth factors are signaling proteins that have important roles in the pathophysiology of SCI. The balance between the proinflammatory and anti-inflammatory effects of these molecules plays a critical role in the progression and outcome of the lesion. The excessive inflammatory Th1 and Th17 phenotypes observed after SCI tilt the scale towards a proinflammatory environment, which exacerbates the deleterious mechanisms present after the injury. These mechanisms include the disruption of the spinal cord blood barrier, edema and ion imbalance, in particular intracellular calcium and sodium concentrations, glutamate excitotoxicity, free radicals, and the inflammatory response contributing to the neurodegenerative process which is characterized by demyelination and apoptosis of neuronal tissue.
Collapse
|
21
|
Moidunny S, Matos M, Wesseling E, Banerjee S, Volsky DJ, Cunha RA, Agostinho P, Boddeke HW, Roy S. Oncostatin M promotes excitotoxicity by inhibiting glutamate uptake in astrocytes: implications in HIV-associated neurotoxicity. J Neuroinflammation 2016; 13:144. [PMID: 27287400 PMCID: PMC4903004 DOI: 10.1186/s12974-016-0613-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 06/06/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Elevated levels of oncostatin M (OSM), an interleukin-6 cytokine family member, have been observed in HIV-1-associated neurocognitive disorders (HAND) and Alzheimer's disease. However, the function of OSM in these disease conditions is unclear. Since deficient glutamate uptake by astrocytes is instrumental in HAND-associated neurotoxicity, we hypothesized that OSM impairs glutamate uptake in astrocytes and thereby promotes neuronal excitotoxicity. METHODS Primary cultures of mouse cortical astrocytes, neurons, microglia, and BV2 cell line were used. The expression of glutamate transporters (GLAST/EAAT1 and GLT-1/EAAT2) was investigated using real-time PCR and Western blot, and their activity was assessed by measuring (3)H-D-aspartate uptake. Neuronal toxicity was measured using the colorimetric MTT (3-(4,5-dimethylthiazol-2-yl-) 2,5-diphenyltetrazolium bromide) assay and immunocytochemistry. A chimeric HIV-1 that infects murine cells (EcoHIV/NL4-3-GFP virus (EcoHIV)) was used to investigate whether the virus induces OSM, OSM receptor (OSMR)-β, glycoprotein 130 (gp130), GLT-1, GLAST (mRNA and protein), and OSM release (ELISA) in cultured BV2 cells, primary microglia, or astrocytes. Statistical analyses of the data were performed using one-way ANOVA (to allow multiple comparisons) and two-tailed Student's t test. RESULTS OSM treatment (10 ng/mL) time-dependently reduced GLAST and GLT-1 expression and inhibited (3)H-D-aspartate uptake in cultured astrocytes in a concentration-dependent manner, an effect prevented by the Janus kinase (JAK)/signal transducers and activators of transcription (STAT)3 inhibitor AG490. Down-regulation of astrocytic glutamate transport by OSM resulted in NMDA receptor-dependent excitotoxicity in cortical neurons. Infection with EcoHIV induced OSM gene expression and protein release in BV2 cells and microglia, but not in astrocytes. Conversely, EcoHIV caused a fivefold increase in OSMR-β mRNA (but not gp130) and protein in astrocytes, but not in microglia, which did not express OSMR-β protein. Finally, astrocytic expression of GLAST gene was unaffected by EcoHIV, whereas GLT-1 mRNA was increased by twofold. CONCLUSIONS We provide first evidence that activation of JAK/STAT3 signaling by OSM inhibits glutamate uptake in astrocytes, which results in neuronal excitotoxicity. Our findings with EcoHIV suggest that targeting OSMR-β signaling in astrocytes might alleviate HIV-1-associated excitotoxicity.
Collapse
Affiliation(s)
- Shamsudheen Moidunny
- Department of Surgery, Division of Basic and Translational Research, University of Minnesota, Minneapolis, MN, USA
| | - Marco Matos
- Center for Neuroscience of Coimbra, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Evelyn Wesseling
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Santanu Banerjee
- Department of Surgery, Division of Basic and Translational Research, University of Minnesota, Minneapolis, MN, USA
| | - David J Volsky
- Molecular Virology Division, St. Luke's-Roosevelt Hospital Center, New York, USA
| | - Rodrigo A Cunha
- Center for Neuroscience of Coimbra, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paula Agostinho
- Center for Neuroscience of Coimbra, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Hendrikus W Boddeke
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sabita Roy
- Department of Surgery, Division of Basic and Translational Research, University of Minnesota, Minneapolis, MN, USA. .,Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
22
|
Ito K, Sanosaka T, Igarashi K, Ideta-Otsuka M, Aizawa A, Uosaki Y, Noguchi A, Arakawa H, Nakashima K, Takizawa T. Identification of genes associated with the astrocyte-specific gene Gfap during astrocyte differentiation. Sci Rep 2016; 6:23903. [PMID: 27041678 PMCID: PMC4819225 DOI: 10.1038/srep23903] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/16/2016] [Indexed: 01/15/2023] Open
Abstract
Chromosomes and genes are non-randomly arranged within the mammalian cell nucleus, and gene clustering is of great significance in transcriptional regulation. However, the relevance of gene clustering and their expression during the differentiation of neural precursor cells (NPCs) into astrocytes remains unclear. We performed a genome-wide enhanced circular chromosomal conformation capture (e4C) to screen for genes associated with the astrocyte-specific gene glial fibrillary acidic protein (Gfap) during astrocyte differentiation. We identified 18 genes that were specifically associated with Gfap and expressed in NPC-derived astrocytes. Our results provide additional evidence for the functional significance of gene clustering in transcriptional regulation during NPC differentiation.
Collapse
Affiliation(s)
- Kenji Ito
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Tsukasa Sanosaka
- Stem Cell Biology and Medicine, Department of Stem cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Katsuhide Igarashi
- Life Science Tokyo Advanced Research Center (L-StaR), Pharmacy and Pharmaceutical Science, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-5801, Japan
| | - Maky Ideta-Otsuka
- Life Science Tokyo Advanced Research Center (L-StaR), Pharmacy and Pharmaceutical Science, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-5801, Japan
| | - Akira Aizawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yuichi Uosaki
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Azumi Noguchi
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hirokazu Arakawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Kinichi Nakashima
- Stem Cell Biology and Medicine, Department of Stem cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takumi Takizawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
23
|
Irudayam JI, Contreras D, Spurka L, Subramanian A, Allen J, Ren S, Kanagavel V, Nguyen Q, Ramaiah A, Ramamoorthy K, French SW, Klein AS, Funari V, Arumugaswami V. Characterization of type I interferon pathway during hepatic differentiation of human pluripotent stem cells and hepatitis C virus infection. Stem Cell Res 2015; 15:354-364. [PMID: 26313525 DOI: 10.1016/j.scr.2015.08.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 06/26/2015] [Accepted: 08/13/2015] [Indexed: 01/08/2023] Open
Abstract
Pluripotent stem cells are being actively studied as a cell source for regenerating damaged liver. For long-term survival of engrafting cells in the body, not only do the cells have to execute liver-specific function but also withstand the physical strains and invading pathogens. The cellular innate immune system orchestrated by the interferon (IFN) pathway provides the first line of defense against pathogens. The objective of this study is to assess the innate immune function as well as to systematically profile the IFN-induced genes during hepatic differentiation of pluripotent stem cells. To address this objective, we derived endodermal cells (day 5 post-differentiation), hepatoblast (day 15) and hepatocyte-like cells (day 21) from human embryonic stem cells (hESCs). Day 5, 15 and 21 cells were stimulated with IFN-α and subjected to IFN pathway analysis. Transcriptome analysis was carried out by RNA sequencing. The results showed that the IFN-α treatment activated STAT-JAK pathway in differentiating cells. Transcriptome analysis indicated stage specific expression of classical and non-classical IFN-stimulated genes (ISGs). Subsequent validation confirmed the expression of novel ISGs including RASGRP3, CLMP and TRANK1 by differentiated hepatic cells upon IFN treatment. Hepatitis C virus replication in hESC-derived hepatic cells induced the expression of ISGs--LAMP3, ETV7, RASGRP3, and TRANK1. The hESC-derived hepatic cells contain intact innate system and can recognize invading pathogens. Besides assessing the tissue-specific functions for cell therapy applications, it may also be important to test the innate immune function of engrafting cells to ensure adequate defense against infections and improve graft survival.
Collapse
Affiliation(s)
- Joseph Ignatius Irudayam
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Deisy Contreras
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lindsay Spurka
- Cedars-Sinai Genomics Core, Medical Genetics Institute, Cedars-Sinai Medical Center Los Angeles, CA 90048, USA
| | - Aparna Subramanian
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jenieke Allen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Songyang Ren
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Vidhya Kanagavel
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Quoclinh Nguyen
- Cedars-Sinai Genomics Core, Medical Genetics Institute, Cedars-Sinai Medical Center Los Angeles, CA 90048, USA
| | - Arunachalam Ramaiah
- Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, Karnataka 560012, India.,Hindustan Genomics Institute, SVA Medical Center, Kadayam, Tamil Nadu 627415, India
| | - Kalidas Ramamoorthy
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu 627012, India.,Hindustan Genomics Institute, SVA Medical Center, Kadayam, Tamil Nadu 627415, India
| | - Samuel W French
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles CA 90095, USA
| | - Andrew S Klein
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Surgery, University of California at Los Angeles, Los Angeles CA 90095, USA
| | - Vincent Funari
- Cedars-Sinai Genomics Core, Medical Genetics Institute, Cedars-Sinai Medical Center Los Angeles, CA 90048, USA
| | - Vaithilingaraja Arumugaswami
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.,Department of Surgery, University of California at Los Angeles, Los Angeles CA 90095, USA
| |
Collapse
|
24
|
Nasi S, So A, Combes C, Daudon M, Busso N. Interleukin-6 and chondrocyte mineralisation act in tandem to promote experimental osteoarthritis. Ann Rheum Dis 2015; 75:1372-9. [PMID: 26253096 DOI: 10.1136/annrheumdis-2015-207487] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/14/2015] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Basic calcium phosphate (BCP) crystal and interleukin 6 (IL-6) have been implicated in osteoarthritis (OA). We hypothesise that these two factors may be linked in a reciprocal amplification loop which leads to OA. METHODS Primary murine chondrocytes and human cartilage explants were incubated with hydroxyapatite (HA) crystals, a form of BCP, and the modulation of cytokines and matrix-degrading enzymes assayed. The ability of IL-6 to stimulate chondrocyte calcification was assessed in vitro. The mechanisms underlying the effects of HA on chondrocytes were investigated using chemical inhibitors, and the pathways mediating IL-6-induced calcification characterised by quantifying the expression of genes involved in chondrocyte mineralisation. The role of calcification in vivo was studied in the meniscectomy model of murine OA (MNX), and the link between IL-6 and cartilage degradation investigated by histology. RESULTS In chondrocytes, BCP crystals stimulated IL-6 secretion, further amplified in an autocrine loop, through signalling pathways involving Syk and PI3 kinases, Jak2 and Stat3 molecules. Exogenous IL-6 promoted calcium-containing crystal formation and upregulation of genes involved in calcification: the pyrophosphate channel Ank, the calcium channel Annexin5 and the sodium/phosphate cotransporter Pit-1. Treatment of chondrocytes with IL-6 inhibitors significantly inhibited IL-6-induced crystal formation. In meniscectomised mice, increasing deposits of BCP crystals were observed around the joint and correlated with cartilage degradation and IL-6 expression. Finally, BCP crystals induced proteoglycan loss and IL-6 expression in human cartilage explants, which were reduced by an IL-6 inhibitor. CONCLUSIONS BCP crystals and IL-6 form a positive feedback loop leading to OA. Targeting calcium-containing crystal formation and/or IL-6 are promising therapeutic strategies in OA.
Collapse
Affiliation(s)
- Sonia Nasi
- Service of Rheumatology, Department of Musculoskeletal Medicine, CHUV and University of Lausanne, Lausanne, Switzerland
| | - Alexander So
- Service of Rheumatology, Department of Musculoskeletal Medicine, CHUV and University of Lausanne, Lausanne, Switzerland
| | - Christèle Combes
- CIRIMAT, UMR 5085 INPT-UPS-CNRS, Université de Toulouse, ENSIACET, Toulouse, France
| | - Michel Daudon
- AP-HP, service d'Explorations Fonctionnelles, hôpital Tenon, Paris, France
| | - Nathalie Busso
- Service of Rheumatology, Department of Musculoskeletal Medicine, CHUV and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
25
|
Janssens K, Maheshwari A, Van den Haute C, Baekelandt V, Stinissen P, Hendriks JJA, Slaets H, Hellings N. Oncostatin M protects against demyelination by inducing a protective microglial phenotype. Glia 2015; 63:1729-37. [DOI: 10.1002/glia.22840] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/01/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Kris Janssens
- Department of Immunology; Biomedical Research Institute, Hasselt University; Diepenbeek Belgium
| | - Anurag Maheshwari
- Department of Immunology; Biomedical Research Institute, Hasselt University; Diepenbeek Belgium
| | - Chris Van den Haute
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy; KU Leuven; Kapucijnenvoer 33 Leuven Belgium
- Department of Neurosciences and Department of Pharmaceutical and Pharmacological Sciences; Leuven Viral Vector Core, KU Leuven; Kapucijnenvoer 33 Leuven Belgium
| | - Veerle Baekelandt
- Department of Neurosciences, Laboratory for Neurobiology and Gene Therapy; KU Leuven; Kapucijnenvoer 33 Leuven Belgium
| | - Piet Stinissen
- Department of Immunology; Biomedical Research Institute, Hasselt University; Diepenbeek Belgium
| | - Jerome J. A. Hendriks
- Department of Immunology; Biomedical Research Institute, Hasselt University; Diepenbeek Belgium
| | - Helena Slaets
- Department of Immunology; Biomedical Research Institute, Hasselt University; Diepenbeek Belgium
| | - Niels Hellings
- Department of Immunology; Biomedical Research Institute, Hasselt University; Diepenbeek Belgium
| |
Collapse
|
26
|
Rodrigues SF, Granger DN. Blood cells and endothelial barrier function. Tissue Barriers 2015; 3:e978720. [PMID: 25838983 DOI: 10.4161/21688370.2014.978720] [Citation(s) in RCA: 205] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022] Open
Abstract
The barrier properties of endothelial cells are critical for the maintenance of water and protein balance between the intravascular and extravascular compartments. An impairment of endothelial barrier function has been implicated in the genesis and/or progression of a variety of pathological conditions, including pulmonary edema, ischemic stroke, neurodegenerative disorders, angioedema, sepsis and cancer. The altered barrier function in these conditions is often linked to the release of soluble mediators from resident cells (e.g., mast cells, macrophages) and/or recruited blood cells. The interaction of the mediators with receptors expressed on the surface of endothelial cells diminishes barrier function either by altering the expression of adhesive proteins in the inter-endothelial junctions, by altering the organization of the cytoskeleton, or both. Reactive oxygen species (ROS), proteolytic enzymes (e.g., matrix metalloproteinase, elastase), oncostatin M, and VEGF are part of a long list of mediators that have been implicated in endothelial barrier failure. In this review, we address the role of blood borne cells, including, neutrophils, lymphocytes, monocytes, and platelets, in the regulation of endothelial barrier function in health and disease. Attention is also devoted to new targets for therapeutic intervention in disease states with morbidity and mortality related to endothelial barrier dysfunction.
Collapse
Key Words
- AJ, Adherens junctions
- ANG-1, Angiopoietin 1
- AQP, Aquaporins
- BBB, blood brain barrier
- CNS, Central nervous system
- COPD, Chronic obstructive pulmonary disease
- EAE, Experimental autoimmune encephalomyelitis
- EPAC1, Exchange protein activated by cyclic AMP
- ERK1/2, Extracellular signal-regulated kinases 1 and 2
- Endothelial barrier
- FA, Focal adhesions
- FAK, focal adhesion tyrosine kinase
- FoxO1, Forkhead box O1
- GAG, Glycosaminoglycans
- GDNF, Glial cell-derived neurotrophic factor
- GJ, Gap junctions
- GPCR, G-protein coupled receptors
- GTPase, Guanosine 5'-triphosphatase
- HMGB-1, High mobility group box 1
- HRAS, Harvey rat sarcoma viral oncogene homolog
- ICAM-1, Intercellular adhesion molecule 1
- IL-1β, Interleukin 1 beta
- IP3, Inositol 1,4,5-triphosphate
- JAM, Junctional adhesion molecules
- MEK, Mitogen-activated protein kinase kinase
- MLC, Myosin light chain
- MLCK, Myosin light-chain kinase
- MMP, Matrix metalloproteinases
- NO, Nitric oxide
- OSM, Oncostatin M
- PAF, Platelet activating factor
- PDE, Phosphodiesterase
- PKA, Protein kinase A
- PNA, Platelet-neutrophil aggregates
- ROS, Reactive oxygen species
- Rac1, Ras-related C3 botulinum toxin substrate 1
- Rap1, Ras-related protein 1
- RhoA, Ras homolog gene family, member A
- S1P, Sphingosine-1-phosphate
- SCID, Severe combined immunodeficient
- SOCS-3, Suppressors of cytokine signaling 3
- Shp-2, Src homology 2 domain-containing phosphatase 2
- Src, Sarcoma family of protein kinases
- TEER, Transendothelial electrical resistance
- TGF-beta1, Transforming growth factor-beta1
- TJ, Tight junctions
- TNF-, Tumor necrosis factor alpha
- VCAM-1, Vascular cell adhesion molecule 1
- VE, Vascular endothelial
- VE-PTP, Vascular endothelial receptor protein tyrosine phosphatase
- VEGF, Vascular endothelial growth factor
- VVO, Vesiculo-vacuolar organelle
- ZO, Zonula occludens
- cAMP, 3'-5'-cyclic adenosine monophosphate
- erythrocytes
- leukocytes
- pSrc, Phosphorylated Src
- platelets
- vascular permeability
Collapse
Affiliation(s)
- Stephen F Rodrigues
- Department of Clinical and Toxicological Analyses; School of Pharmaceutical Sciences; University of Sao Paulo ; Sao Paulo, Brazil
| | - D Neil Granger
- Department of Molecular and Cellular Physiology; Louisiana State University Health Sciences Center ; Shreveport, LA USA
| |
Collapse
|
27
|
PERK-dependent activation of JAK1 and STAT3 contributes to endoplasmic reticulum stress-induced inflammation. Mol Cell Biol 2014; 34:3911-25. [PMID: 25113558 DOI: 10.1128/mcb.00980-14] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Neuroinflammation and endoplasmic reticulum (ER) stress are associated with many neurological diseases. Here, we have examined the interaction between ER stress and JAK/STAT-dependent inflammation in glial cells. We show that ER stress is present in the central nervous system (CNS) concomitant with inflammation and astrogliosis in the multiple sclerosis (MS) mouse model of experimental autoimmune encephalomyelitis (EAE). Astrocytes do not easily succumb to ER stress but rather activate an inflammatory program involving activation of STAT3 in a JAK1-dependent fashion. ER stress-induced activation of the JAK1/STAT3 axis leads to expression of interleukin 6 (IL-6) and several chemokines. Moreover, the activation of STAT3 signaling is dependent on PERK, a central component of the ER stress response, which we show is phosphorylated by JAK1. Disruption of PERK abrogates ER stress-induced activation of STAT3 and subsequent gene expression. Additionally, ER-stressed astrocytes, via paracrine signaling, can stimulate activation of microglia, leading to production of IL-6 and oncostatin M (OSM). These IL-6 cytokines can then synergize with ER stress in astrocytes to drive inflammation. Together, this work describes a new PERK/JAK1/STAT3 signaling pathway that elicits a feed-forward inflammatory loop involving astrocytes and microglia to drive neuroinflammation, which may be relevant in diseases such as MS.
Collapse
|
28
|
Hsu MP, Frausto R, Rose-John S, Campbell IL. Analysis of IL-6/gp130 family receptor expression reveals that in contrast to astroglia, microglia lack the oncostatin M receptor and functional responses to oncostatin M. Glia 2014; 63:132-41. [DOI: 10.1002/glia.22739] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 07/24/2014] [Indexed: 02/04/2023]
Affiliation(s)
- Meng-Ping Hsu
- School of Molecular Bioscience and the Bosch Institute, University of Sydney; Sydney NSW Australia
| | - Ricardo Frausto
- School of Molecular Bioscience and the Bosch Institute, University of Sydney; Sydney NSW Australia
| | - Stefan Rose-John
- Department of Biochemistry; Christian-Albrechts University of Kiel; Kiel Germany
| | - Iain L. Campbell
- School of Molecular Bioscience and the Bosch Institute, University of Sydney; Sydney NSW Australia
| |
Collapse
|
29
|
Robson MJ, Turner RC, Naser ZJ, McCurdy CR, O'Callaghan JP, Huber JD, Matsumoto RR. SN79, a sigma receptor antagonist, attenuates methamphetamine-induced astrogliosis through a blockade of OSMR/gp130 signaling and STAT3 phosphorylation. Exp Neurol 2014; 254:180-9. [PMID: 24508558 DOI: 10.1016/j.expneurol.2014.01.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Revised: 01/22/2014] [Accepted: 01/27/2014] [Indexed: 12/11/2022]
Abstract
Methamphetamine (METH) exposure results in dopaminergic neurotoxicity in striatal regions of the brain, an effect that has been linked to an increased risk of Parkinson's disease. Various aspects of neuroinflammation, including astrogliosis, are believed to be contributory factors in METH neurotoxicity. METH interacts with sigma receptors at physiologically relevant concentrations and treatment with sigma receptor antagonists has been shown to mitigate METH-induced neurotoxicity in rodent models. Whether these compounds alter the responses of glial cells within the central nervous system to METH however has yet to be determined. Therefore, the purpose of the current study was to determine whether the sigma receptor antagonist, SN79, mitigates METH-induced striatal reactive astrogliosis. Male, Swiss Webster mice treated with a neurotoxic regimen of METH exhibited time-dependent increases in striatal gfap mRNA and concomitant increases in GFAP protein, indicative of astrogliosis. This is the first report that similar to other neurotoxicants that induce astrogliosis through the activation of JAK2/STAT3 signaling by stimulating gp-130-linked cytokine signaling resulting from neuroinflammation, METH treatment also increases astrocytic oncostatin m receptor (OSMR) expression and the phosphorylation of STAT3 (Tyr-705) in vivo. Pretreatment with SN79 blocked METH-induced increases in OSMR, STAT3 phosphorylation and astrocyte activation within the striatum. Additionally, METH treatment resulted in striatal cellular degeneration as measured by Fluoro-Jade B, an effect that was mitigated by SN79. The current study provides evidence that sigma receptor antagonists attenuate METH-induced astrocyte activation through a pathway believed to be shared by various neurotoxicants.
Collapse
Affiliation(s)
- Matthew J Robson
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, 1 Medical Center Dr., West Virginia University Health Sciences Center, Morgantown, WV 26506, USA; Department of Pharmacology, School of Medicine, Vanderbilt University, 1161 21st Ave S., Nashville, TN 37232, USA
| | - Ryan C Turner
- Department of Neurosurgery, School of Medicine, West Virginia University, 1 Medical Center Dr., West Virginia University Health Sciences Center, Morgantown, WV 26506, USA; Center for Neuroscience, School of Medicine, West Virginia University, 1 Medical Center Dr., West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - Zachary J Naser
- Department of Neurosurgery, School of Medicine, West Virginia University, 1 Medical Center Dr., West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, School of Pharmacy, University of Mississippi, P.O. Box 1848, University, MS 38677-1848, USA; Department of Pharmacology, School of Pharmacy, University of Mississippi, P.O. Box 1848, University, MS 38677-1848, USA
| | - James P O'Callaghan
- Health Effects Laboratory Division, Centers for Disease Control and Prevention, National Institute of Occupational Safety and Health, 1095 Willowdale Rd., Morgantown, WV 26505, USA
| | - Jason D Huber
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, 1 Medical Center Dr., West Virginia University Health Sciences Center, Morgantown, WV 26506, USA; Center for Neuroscience, School of Medicine, West Virginia University, 1 Medical Center Dr., West Virginia University Health Sciences Center, Morgantown, WV 26506, USA
| | - Rae R Matsumoto
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, West Virginia University, 1 Medical Center Dr., West Virginia University Health Sciences Center, Morgantown, WV 26506, USA; Center for Neuroscience, School of Medicine, West Virginia University, 1 Medical Center Dr., West Virginia University Health Sciences Center, Morgantown, WV 26506, USA.
| |
Collapse
|
30
|
Richards CD. The enigmatic cytokine oncostatin m and roles in disease. ISRN INFLAMMATION 2013; 2013:512103. [PMID: 24381786 PMCID: PMC3870656 DOI: 10.1155/2013/512103] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 09/29/2013] [Indexed: 12/11/2022]
Abstract
Oncostatin M is a secreted cytokine involved in homeostasis and in diseases involving chronic inflammation. It is a member of the gp130 family of cytokines that have pleiotropic functions in differentiation, cell proliferation, and hematopoetic, immunologic, and inflammatory networks. However, Oncostatin M also has activities novel to mediators of this cytokine family and others and may have fundamental roles in mechanisms of inflammation in pathology. Studies have explored Oncostatin M functions in cancer, bone metabolism, liver regeneration, and conditions with chronic inflammation including rheumatoid arthritis, lung and skin inflammatory disease, atherosclerosis, and cardiovascular disease. This paper will review Oncostatin M biology in a historical fashion and focus on its unique activities, in vitro and in vivo, that differentiate it from other cytokines and inspire further study or consideration in therapeutic approaches.
Collapse
Affiliation(s)
- Carl D. Richards
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, 1280 Main Street, West, Hamilton, ON, Canada L8S 4K1
| |
Collapse
|
31
|
Dey G, Radhakrishnan A, Syed N, Thomas JK, Nadig A, Srikumar K, Mathur PP, Pandey A, Lin SK, Raju R, Prasad TSK. Signaling network of Oncostatin M pathway. J Cell Commun Signal 2013; 7:103-8. [PMID: 23255051 PMCID: PMC3660694 DOI: 10.1007/s12079-012-0186-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 12/03/2012] [Indexed: 12/23/2022] Open
Affiliation(s)
- Gourav Dey
- />Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, 560 066 India
| | - Aneesha Radhakrishnan
- />Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, 560 066 India
- />Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, 605 014 India
| | - Nazia Syed
- />Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, 560 066 India
- />Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, 605 014 India
| | - Joji Kurian Thomas
- />Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, 560 066 India
- />School of Biotechnology, Amrita Vishwa Vidhyapeetam, Kollam, 690 525 India
| | - Arpitha Nadig
- />Department of Bioinformatics, Kuvempu University, Karnataka Shankaraghatta, 577 451 India
| | - Kotteazeth Srikumar
- />Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, 605 014 India
| | - Premendu Prakash Mathur
- />Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Puducherry, 605 014 India
- />KIIT University, Bhubaneswar, 751 024 India
| | - Akhilesh Pandey
- />McKusick-Nathans Institute of Genetic Medicine and Departments of Biological Chemistry, Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Sze-Kwan Lin
- />School of Dentistry, College of Medicine, National Taiwan University, 1 Chang-Te Street, Taipei, 10016 Taiwan
| | - Rajesh Raju
- />Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, 560 066 India
| | - T. S. Keshava Prasad
- />Institute of Bioinformatics, International Tech Park, Whitefield, Bangalore, 560 066 India
- />School of Biotechnology, Amrita Vishwa Vidhyapeetam, Kollam, 690 525 India
| |
Collapse
|
32
|
Astrocytes protect neurons against methylmercury via ATP/P2Y(1) receptor-mediated pathways in astrocytes. PLoS One 2013; 8:e57898. [PMID: 23469098 PMCID: PMC3585279 DOI: 10.1371/journal.pone.0057898] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 01/27/2013] [Indexed: 11/19/2022] Open
Abstract
Methylmercury (MeHg) is a well known environmental pollutant that induces serious neuronal damage. Although MeHg readily crosses the blood-brain barrier, and should affect both neurons and glial cells, how it affects glia or neuron-to-glia interactions has received only limited attention. Here, we report that MeHg triggers ATP/P2Y1 receptor signals in astrocytes, thereby protecting neurons against MeHg via interleukin-6 (IL-6)-mediated pathways. MeHg increased several mRNAs in astrocytes, among which IL-6 was the highest. For this, ATP/P2Y1 receptor-mediated mechanisms were required because the IL-6 production was (i) inhibited by a P2Y1 receptor antagonist, MRS2179, (ii) abolished in astrocytes obtained from P2Y1 receptor-knockout mice, and (iii) mimicked by exogenously applied ATP. In addition, (iv) MeHg released ATP by exocytosis from astrocytes. As for the intracellular mechanisms responsible for IL-6 production, p38 MAP kinase was involved. MeHg-treated astrocyte-conditioned medium (ACM) showed neuro-protective effects against MeHg, which was blocked by anti-IL-6 antibody and was mimicked by the application of recombinant IL-6. As for the mechanism of neuro-protection by IL-6, an adenosine A1 receptor-mediated pathway in neurons seems to be involved. Taken together, when astrocytes sense MeHg, they release ATP that autostimulates P2Y1 receptors to upregulate IL-6, thereby leading to A1 receptor-mediated neuro-protection against MeHg.
Collapse
|
33
|
Dumas A, Lagarde S, Laflamme C, Pouliot M. Oncostatin M decreases interleukin-1 β secretion by human synovial fibroblasts and attenuates an acute inflammatory reaction in vivo. J Cell Mol Med 2012; 16:1274-85. [PMID: 21854541 PMCID: PMC3823080 DOI: 10.1111/j.1582-4934.2011.01412.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Oncostatin M (OSM) is a pleiotropic cytokine of the IL-6 family and displays both pro-inflammatory and anti-inflammatory activities. We studied the impact of OSM on the gene activation profile of human synovial cells, which play a central role in the progression of inflammatory responses in joints. In synovial cells stimulated with lipopolysaccharide and recombinant human granulocyte-macrophage colony-stimulating factor, recombinant human OSM and native OSM secreted by human granulocytes both reduced the gene expression and secretion of IL-1β and CXCL8, but increased that of IL-6 and CCL2. This impact on synovial cell activation was not obtained using IL-6 or leukaemia inhibitory factor. Signal transducer and activator of transcription-1 appeared to mediate the effects of OSM on stimulated human synovial fibroblasts. In the murine dorsal air pouch model of inflammation, OSM reduced the expression of the pro-inflammatory cytokines IL-1β and TNF-α in lining tissues, and their presence in the cavity. These results as a whole suggest an anti-inflammatory role for OSM, guiding inflammatory processes towards resolution.
Collapse
Affiliation(s)
- Aline Dumas
- Centre de Recherche en Rhumatologie et Immunologie du CHUQ, and Department of Microbiology-Infectiology and Immunology, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| | | | | | | |
Collapse
|
34
|
Schulz I, Engel C, Niestroj AJ, Zeitschel U, Menge K, Kehlen A, Meyer A, Rossner S, Demuth HU. Heteroarylketones inhibit astroglial interleukin-6 expression via a STAT3/NF-κB signaling pathway. J Neuroinflammation 2011; 8:86. [PMID: 21801384 PMCID: PMC3161871 DOI: 10.1186/1742-2094-8-86] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 07/29/2011] [Indexed: 01/30/2023] Open
Abstract
Background Elevated brain levels of the pleiotropic cytokine interleukin-6, which is mainly secreted from activated local astrocytes, contribute to pathological events including neuroinflammation and neurodegeneration. Thus, inhibition of pathological IL-6 expression provides a rationale strategy for targeting the onset or further progression of neurological disorders including Alzheimer's disease, multiple sclerosis, Parkinson's disease and traumatic brain injury. The purpose of this study was to identify and to characterize new potent inhibitors of astrocytic IL-6 expression for further therapeutic development of novel anti-inflammatory and neuroprotective drugs. Methods Oncostatin M (OSM)-treated human glioma U343 cells were used as model for induction of astrocytic IL-6 expression. This model was characterized by immunoblotting, siRNA technique, ELISA and qRT-PCR and used to screen low molecular weight compound libraries for IL-6-lowering effects. To validate bioactive compounds identified from library screens, bacterial lipopolysaccharide was used to induce IL-6 expression in cultivated primary astrocytes and in mice in vivo. To dissect underlying molecular mechanisms, protein extracts from OSM-treated U343 cells were analyzed by phospho-specific immunoblotting and immunocytochemistry as well as by co-immunoprecipitation. Results OSM-treatment (100 ng/ml; 24 h) led to 30-fold increase of IL-6 secretion from U343 cells. The temporal profile of IL-6 mRNA induction displayed a biphasic induction pattern with peak synthesis at 1 h (6.5-fold) and 16 h (5.5-fold) post stimulation. IL-6 protein release did not show that biphasic pattern and was detected as early as 3 h post stimulation reaching a maximum at 24 h. The screen of compound libraries identified a set of heteroarylketones (HAKs) as potent inhibitors of IL-6 secretion. HAK compounds affected the second peak in IL-6 mRNA synthesis, whereas the first peak was insensitive to HAK treatment. HAK compounds also suppressed lipopolysaccharide-induced IL-6 expression in primary murine astrocytes as well as in brain and plasma samples from lipopolysaccharide-treated mice. Finally, HAK compounds were demonstrated to specifically suppress the OSM-induced phosphorylation of STAT3 at serine 727 and the physical interaction of pSTAT3S727 with p65. Conclusion Heteroarylketone compounds are potent inhibitors of IL-6 expression in vitro and in vivo and may represent a new class of potent anti-inflammatory and neuroprotective drugs.
Collapse
Affiliation(s)
- Ingo Schulz
- Probiodrug AG, Weinbergweg 22, Halle/Saale, 06120, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Glezer I, Rivest S. Oncostatin M is a novel glucocorticoid-dependent neuroinflammatory factor that enhances oligodendrocyte precursor cell activity in demyelinated sites. Brain Behav Immun 2010; 24:695-704. [PMID: 20083191 DOI: 10.1016/j.bbi.2010.01.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 01/12/2010] [Accepted: 01/13/2010] [Indexed: 10/20/2022] Open
Abstract
The innate immune reaction to tissue injury is a natural process, which can have detrimental effects in the absence of negative feedbacks by glucocorticoids (GCs). Although acute lipopolysaccharide (LPS) challenge is relatively harmless to the brain parenchyma of adult animals, the endotoxin is highly neurotoxic in animals that are treated with the GC receptor antagonist RU486. This study investigated the role of cytokines of the gp130-related family in these effects, because they are essential components of the inflammatory process that provide survival signals to neurons. Intracerebral LPS injection stimulated expression of several members of this family of cytokines, but oncostatin M (Osm) was the unique ligand to be completely inhibited by the RU486 treatment. OSM receptor (Osmr) is expressed mainly in astrocytes and endothelial cells following LPS administration and GCs are directly responsible for its transcriptional activation in the presence of the endotoxin. In a mouse model of demyelination, exogenous OSM significantly modulated the expression of genes involved in the mobilization of oligodendrocyte precursor cells (OPCs), differentiation of oligodendrocyte, and production of myelin. In conclusion, the activation of OSM signaling is a mechanism activated by TLR4 in the presence of negative feedback by GCs on the innate immune system of the brain. OSM absence is associated with detrimental effects of LPS, whereas exogenous OSM favors repair response to demyelinated regions.
Collapse
Affiliation(s)
- Isaias Glezer
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
36
|
Williams R, Yao H, Dhillon NK, Buch SJ. HIV-1 Tat co-operates with IFN-gamma and TNF-alpha to increase CXCL10 in human astrocytes. PLoS One 2009; 4:e5709. [PMID: 19479051 PMCID: PMC2684622 DOI: 10.1371/journal.pone.0005709] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 05/01/2009] [Indexed: 12/12/2022] Open
Abstract
HIV-associated neurological disorders (HAND) are estimated to affect 60% of the HIV infected population. HIV-encephalitis (HIVE), the pathological correlate of the most severe form of HAND is often characterized by glial activation, cytokine/chemokine dysregulation, and neuronal damage and loss. However, the severity of HIVE correlates better with glial activation rather than viral load. One of the characteristic features of HIVE is the increased amount of the neurotoxic chemokine, CXCL10. This chemokine can be released from astroglia activated with the pro-inflammatory cytokines IFN-γ and TNF-α, in conjunction with HIV-1 Tat, all of which are elevated in HIVE. In an effort to understand the pathogenesis of HAND, this study was aimed at exploring the regulation of CXCL10 by cellular and viral factors during astrocyte activation. Specifically, the data herein demonstrate that the combined actions of HIV-1 Tat and the pro-inflammatory cytokines, IFN-γ and TNF-α, result in the induction of CXCL10 at both the RNA and protein level. Furthermore, CXCL10 induction was found to be regulated transcriptionally by the activation of the p38, Jnk, and Akt signaling pathways and their downstream transcription factors, NF-κB and STAT-1α. Since CXCL10 levels are linked to disease severity, understanding its regulation could aid in the development of therapeutic intervention strategies for HAND.
Collapse
Affiliation(s)
- Rachel Williams
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Honghong Yao
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Navneet K. Dhillon
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Shilpa J. Buch
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail:
| |
Collapse
|
37
|
Baker BJ, Qin H, Benveniste EN. Molecular basis of oncostatin M-induced SOCS-3 expression in astrocytes. Glia 2008; 56:1250-62. [PMID: 18571793 DOI: 10.1002/glia.20694] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Under neuropathological conditions, reactive astrocytes release cytokines and chemokines, which act in an autocrine and/or paracrine fashion to modulate production of immunoregulatory factors from cells including microglia, astrocytes, and neurons. In this way, astrocytes play an important role in orchestrating immune responses within the central nervous system (CNS). Suppressor of cytokine signaling (SOCS) proteins are endogenous, negative regulators of the JAK/STAT signaling pathway and function as attenuators of the immune and inflammatory responses. As such, SOCS proteins may have critical roles in the CNS under neuroinflammatory conditions. In the inflamed CNS, expression of IL-6 cytokine family member oncostatin M (OSM) is elevated; however, its functional effects are not well understood. We demonstrate that OSM is a potent inducer of SOCS-3 in astrocytes. Analysis of the SOCS-3 promoter revealed that an AP-1 element, two IFN-gamma activation sequence (GAS) elements, and a GC-rich region are crucial for SOCS-3 gene expression. Using small interfering RNA against STAT-3, as well as a STAT-3 dominant-negative construct, we demonstrate that STAT-3 activation is critical for OSM induction of SOCS-3 expression. The ERK1/2 and JNK pathways also contribute to OSM-induced SOCS-3 gene expression. OSM stimulation led to a time-dependent recruitment of the transcription factors STAT-3, c-Fos, c-Jun, and Sp1 and the coactivators CREB-binding protein (CBP) and p300 to the endogenous SOCS-3 promoter. These data indicate that OSM-induced activation of STAT-3 and the ERK1/2 and JNK pathways are critical for astrocytic expression of SOCS-3, which provides for feedback inhibition of cytokine-induced inflammatory responses in the CNS.
Collapse
Affiliation(s)
- Brandi J Baker
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0005, USA
| | | | | |
Collapse
|
38
|
Tiffen PG, Omidvar N, Marquez-Almuina N, Croston D, Watson CJ, Clarkson RWE. A dual role for oncostatin M signaling in the differentiation and death of mammary epithelial cells in vivo. Mol Endocrinol 2008; 22:2677-88. [PMID: 18927239 DOI: 10.1210/me.2008-0097] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Recent studies in breast cancer cell lines have shown that oncostatin M (OSM) not only inhibits proliferation but also promotes cell detachment and enhances cell motility. In this study, we have looked at the role of OSM signaling in nontransformed mouse mammary epithelial cells in vitro using the KIM-2 mammary epithelial cell line and in vivo using OSM receptor (OSMR)-deficient mice. OSM and its receptor were up-regulated approximately 2 d after the onset of postlactational mammary regression, in response to leukemia inhibitory factor (LIF)-induced signal transducer and activator of transcription-3 (STAT3). This resulted in sustained STAT3 activity, increased epithelial apoptosis, and enhanced clearance of epithelial structures during the remodeling phase of mammary involution. Concurrently, OSM signaling precipitated the dephosphorylation of STAT5 and repressed expression of the milk protein genes beta-casein and whey acidic protein (WAP). Similarly, during pregnancy, OSM signaling suppressed beta-casein and WAP gene expression. In vitro, OSM but not LIF persistently down-regulated phosphorylated (p)-STAT5, even in the continued presence of prolactin. OSM also promoted the expression of metalloproteinases MMP3, MMP12, and MMP14, which, in vitro, were responsible for OSM-specific apoptosis. Thus, the sequential activation of IL-6-related cytokines during mammary involution culminates in an OSM-dependent repression of epithelial-specific gene expression and the potentiation of epithelial cell extinction mediated, at least in part, by the reciprocal regulation of p-STAT5 and p-STAT3.
Collapse
Affiliation(s)
- Paul G Tiffen
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | | | | | | | | | | |
Collapse
|
39
|
Brantley EC, Benveniste EN. Signal transducer and activator of transcription-3: a molecular hub for signaling pathways in gliomas. Mol Cancer Res 2008; 6:675-84. [PMID: 18505913 DOI: 10.1158/1541-7786.mcr-07-2180] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Glioblastoma is the most common and severe primary brain tumor in adults. Its aggressive and infiltrative nature renders the current therapeutics of surgical resection, radiation, and chemotherapy relatively ineffective. Accordingly, recent research has focused on the elucidation of various signal transduction pathways in glioblastoma, particularly aberrant activation. This review focuses on the signal transducer and activator of transcription-3 (STAT-3) signal transduction pathway in the context of this devastating tumor. STAT-3 is aberrantly activated in human glioblastoma tissues, and this activation is implicated in controlling critical cellular events thought to be involved in gliomagenesis, such as cell cycle progression, apoptosis, angiogenesis, and immune evasion. There are no reports of gain-of-function mutations in glioblastoma; rather, the activation of STAT-3 is thought to be a consequence of either dysregulation of upstream kinases or loss of endogenous inhibitors. This review provides detailed insight into the multiple mechanisms of STAT-3 activation in glioblastoma, as well as describing endogenous and chemical inhibitors of this pathway and their clinical significance. In glioblastoma, STAT-3 acts a molecular hub to link extracellular signals to transcriptional control of proliferation, cell cycle progression, and immune evasion. Because STAT-3 plays this central role in glioblastoma signal transduction, it has significant potential as a therapeutic target.
Collapse
Affiliation(s)
- Emily C Brantley
- Department of Cell Biology, 1918 University Boulevard, MCLM 395A, University of Alabama at Birmingham, Birmingham, AL 35294-0005, USA
| | | |
Collapse
|
40
|
Takata F, Sumi N, Nishioku T, Harada E, Wakigawa T, Shuto H, Yamauchi A, Kataoka Y. Oncostatin M induces functional and structural impairment of blood–brain barriers comprised of rat brain capillary endothelial cells. Neurosci Lett 2008; 441:163-6. [DOI: 10.1016/j.neulet.2008.06.030] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Revised: 06/11/2008] [Accepted: 06/11/2008] [Indexed: 11/24/2022]
|
41
|
Duluc D, Delneste Y, Tan F, Moles MP, Grimaud L, Lenoir J, Preisser L, Anegon I, Catala L, Ifrah N, Descamps P, Gamelin E, Gascan H, Hebbar M, Jeannin P. Tumor-associated leukemia inhibitory factor and IL-6 skew monocyte differentiation into tumor-associated macrophage-like cells. Blood 2007; 110:4319-30. [PMID: 17848619 DOI: 10.1182/blood-2007-02-072587] [Citation(s) in RCA: 339] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Tumor-associated macrophages (TAMs), the most abundant immunosuppressive cells in the tumor microenvironment, originate from blood monocytes and exhibit an IL-10(high)IL-12(low) M2 profile. The factors involved in TAM generation remain unidentified. We identify here leukemia inhibitory factor (LIF) and IL-6 as tumor microenvironmental factors that can promote TAM generation. Ovarian cancer ascites switched monocyte differentiation into TAM-like cells that exhibit most ovarian TAM functional and phenotypic characteristics. Ovarian cancer ascites contained high concentrations of LIF and IL-6. Recombinant LIF and IL-6 skew monocyte differentiation into TAM-like cells by enabling monocytes to consume monocyte-colony-stimulating factor (M-CSF). Depletion of LIF, IL-6, and M-CSF in ovarian cancer ascites suppressed TAM-like cell induction. We extended these observations to different tumor-cell line supernatants. In addition to revealing a new tumor-escape mechanism associated with TAM generation via LIF and IL-6, these findings offer novel therapeutic perspectives to subvert TAM-induced immunosuppression and hence improve T-cell-based antitumor immunotherapy efficacy.
Collapse
|
42
|
Damiani CL, O'Callaghan JP. Recapitulation of cell signaling events associated with astrogliosis using the brain slice preparation. J Neurochem 2007; 100:720-6. [PMID: 17176261 DOI: 10.1111/j.1471-4159.2006.04321.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Astroglial activation constitutes a dominant response to all types of injuries of the CNS. Despite the ubiquitous nature of this cellular reaction to neural injury, a little is known concerning the signaling mechanisms that initiate it. Recently, we demonstrated that astrocytic hypertrophy and enhanced expression of glial fibrillary acidic protein resulting from toxicant-induced neurodegeneration are linked to activation of the janus kinase (JAK)-signal transducer and activator of transcription-3 (STAT3) pathway. These observations implicate ligands at the gp130 receptor as potential upstream effectors of astrogliosis. Here we used the brain slice preparation to examine potential activators of the JAK-STAT3 pathway. Following incubation of freshly cut striatal slices in phosphate-free oxygenated buffer for up to 75 min, we found that slicing the striatum itself was a sufficient stimulus to initiate a rapid activation of the JAK-STAT3 pathway as assessed with immunoblots of pSTAT3((tyr705)) using phospho-state specific antibodies. The mRNA for the gp130 cytokines, leukemia inhibitory factor, interleukin-6 and oncostatin M or the beta-chemokine, monocyte chemoattractive protein (CCl2) also were up-regulated in the slice. Moreover, we could enhance the activation of STAT3((tyr705)) by adding exogenous cytokines to the slice and we could inhibit phosphorylation of STAT3((tyr705)) by addition of tyrosine kinase inhibitors (Lav A and AG490) or neutralizing antibodies directed against leukemia inhibitory factor or oncostatin M. These data suggest that STAT3 activation is an early event in slice-induced glial activation and establishes the brain slice preparation method as a reliable model to examine the signaling mechanisms that underlie glial activation.
Collapse
Affiliation(s)
- Candice L Damiani
- Centers for Disease Control and Prevention, National Institute for Occupational Safety and Health (CDC-NIOSH), Morgantown, WV 26505, USA
| | | |
Collapse
|
43
|
Chen SH, Gillespie GY, Benveniste EN. Divergent effects of oncostatin M on astroglioma cells: influence on cell proliferation, invasion, and expression of matrix metalloproteinases. Glia 2006; 53:191-200. [PMID: 16206166 DOI: 10.1002/glia.20264] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Oncostatin M (OSM), a cytokine of the interleukin-6 (IL-6) family, can either promote or inhibit cell growth in various normal and tumor cells. We addressed the effects of exogenous OSM on the proliferation and invasion of human astroglioma cells. In addition, we investigated one of the possible mechanisms involved: modulation of matrix metalloproteinase (MMP) expression and enzymatic activity. We found that OSM inhibited the proliferation of two human astroglioma cell lines (CH235-MG and U87-MG), and that this effect was not due to apoptosis. The inhibitory effect of OSM on proliferation was mediated through the gp130/OSMRbeta receptor complex. To extend these findings, we analyzed the effects of OSM on primary tumor cells from glioblastoma patients. OSM suppressed the proliferation of primary glioblastoma cells, but not that of normal astrocytes. Interestingly, OSM did not suppress astroglioma cell invasion. This may be due to the differential regulation of MMPs by OSM. We found that OSM inhibited the constitutive expression of MMP-2, while MMP-9 expression was enhanced in astroglioma cell lines. We conclude that OSM inhibits proliferation of human astroglioma cells and primary glioblastoma cells via the gp130/OSMRbeta receptor complex. However, OSM does not affect the invasive capacity of the astroglioma cells, which may be due to the divergent effects of OSM on MMP-2 and MMP-9 expression. Collectively, these findings suggest a complex role for OSM in astroglioma biology.
Collapse
Affiliation(s)
- Shao-Hua Chen
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0005, USA
| | | | | |
Collapse
|
44
|
Godoy-Tundidor S, Cavarretta ITR, Fuchs D, Fiechtl M, Steiner H, Friedbichler K, Bartsch G, Hobisch A, Culig Z. Interleukin-6 and oncostatin M stimulation of proliferation of prostate cancer 22Rv1 cells through the signaling pathways of p38 mitogen-activated protein kinase and phosphatidylinositol 3-kinase. Prostate 2005; 64:209-16. [PMID: 15712220 DOI: 10.1002/pros.20235] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
INTRODUCTION Interleukin-6 (IL-6) is a pleiotropic regulator of prostate cancer cell growth. Oncostatin M (OSM), an IL-6-type cytokine, affects the growth of prostate cancers in a paracrine and autocrine manner. In order to understand better the mechanisms controlling proliferation and intracellular signaling by these cytokines in advanced prostate carcinoma, we performed studies in 22Rv1 cells derived from the relapsed xenograft CWR22R. METHODS Expression of IL-6 and OSM receptors (OSMR-beta) and elements of signal transduction pathways in 22Rv1 cells were investigated by RT-PCR. Proliferation was assessed by cell counting after treatment with either IL-6 or OSM. IL-6 secretion was measured in conditioned medium from 22Rv1 cells by ELISA. Expression and phosphorylation status of signal transducers and activators of transcription factor (STAT) 3, mitogen-activated protein kinases (MAPK) p44/p42 and p38, and protein kinase B (Akt) was investigated by Western blot. RESULTS 22Rv1 cells express both subunits of the IL-6 receptor (gp80 and gp130) and leukemia inhibitory factor receptor-beta (LIFR-beta) but not OSMR-beta. Their proliferation was stimulated by IL-6 or OSM and the maximal effect was observed at a concentration of 10 ng/ml of either cytokine. Interestingly, neither IL-6 nor OSM induced phosphorylation of STAT3. OSM modestly increased the phosphorylation of p38 and both cytokines exerted an effect on Akt phosphorylation. CONCLUSIONS IL-6 and OSM stimulate proliferation of 22Rv1 cells, at least in part through activation of the phosphatidylinositol 3-kinase (PI 3-K) signaling pathway. Our data provide additional evidence for the growth-stimulatory role of IL-6 and related cytokines in advanced prostate cancer and may serve as a basis for the development of novel experimental therapies.
Collapse
|
45
|
Chen SH, Benveniste EN. Oncostatin M: a pleiotropic cytokine in the central nervous system. Cytokine Growth Factor Rev 2005; 15:379-91. [PMID: 15450253 DOI: 10.1016/j.cytogfr.2004.06.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Oncostatin M (OSM), a member of the interleukin-6 (IL-6) cytokine family, has yet to be well studied, especially in the context of the central nervous system (CNS). The biological functions of OSM are complex and variable, depending on the cellular microenvironment. Inflammatory responses and tumor development are among two of the major events that OSM is involved in. Although OSM levels remain low in the normal CNS, elevated expression occurs in pathological conditions. Therefore, it is crucial to understand the regulation of OSM to control its expression and/or its effects. Accumulating data demonstrate that OSM binds to specific receptor complexes, then activates two major signaling pathways: Janus Kinase-Signal Transducers and Activators of Transcription (JAK-STAT) and Mitogen-Activated Protein Kinase (MAPK), to regulate downstream events. In this review, we focus on the biological functions of OSM, the signaling pathways of OSM in the CNS, and OSM involvement in CNS diseases.
Collapse
Affiliation(s)
- Shao-Hua Chen
- Department of Cell Biology, MCLM 386, University of Alabama at Birmingham, 1918 University Boulevard, Birmingham, AL 35294-0005, USA.
| | | |
Collapse
|
46
|
Gu JW, Wang J, Stockton A, Lokitz B, Henegar L, Hall JE. Cytokine gene expression profiles in kidney medulla and cortex of obese hypertensive dogs. Kidney Int 2004; 66:713-21. [PMID: 15253726 DOI: 10.1111/j.1523-1755.2004.00793.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND The molecular mechanisms linking abnormal kidney function and obesity hypertension are poorly understood. This study compared gene expression profiles in the kidney medulla and cortex of obese and lean dogs. METHODS Lean dogs (N= 4) were fed a standard kennel ration and obese dogs (N= 4) were fed the standard diet plus 0.5 to 0.9 kg of cooked beef fat per day for 10 weeks. The dogs were instrumented for continuous monitoring of mean arterial pressure (MAP), heart rate, glomerular filtration rate (GFR), and effective renal plasma flow (RPF). The relative mRNA levels of 375 genes in renal cortex and medulla were determined simultaneously using cDNA membrane arrays (R&D Systems). RESULTS The high fat diet increased body weight by 57% and MAP increased by 24 mm Hg (112 +/- 1 mm Hg vs. 88 +/- 3 mm Hg) in obese compared to lean dogs. In obese dogs, expression of 11 and 13 genes changed significantly (N= 4; P < 0.05) in the renal medulla and the cortex, respectively, relative to the lean dogs. Differences in renal gene expression profiles between lean and obese dogs were closely related to functional pathways, including those associated with sympathetic activation, inflammatory response, matrix formation, angiogenesis, endothelial dysfunction, attenuated actions of leptin, and attenuated cell survival. CONCLUSION A high fat diet in dogs is associated with marked changes in renal gene expression profiles that provide potential molecular links to pathways associated with altered renal function and structure in obesity hypertension.
Collapse
Affiliation(s)
- Jian-Wei Gu
- Department of Physiology & Biophysics, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Tong L, Smyth D, Kerr C, Catterall J, Richards CD. Mitogen-activated protein kinases Erk1/2 and p38 are required for maximal regulation of TIMP-1 by oncostatin M in murine fibroblasts. Cell Signal 2004; 16:1123-32. [PMID: 15240007 DOI: 10.1016/j.cellsig.2004.03.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2004] [Revised: 03/02/2004] [Accepted: 03/03/2004] [Indexed: 11/17/2022]
Abstract
Oncostatin M (OSM) regulates expression of various genes in connective tissue (CT) cells, including tissue inhibitor of metalloproteinases-1 (TIMP-1). In mouse fibroblast cell lines MLg, NIH 3T3 and primary mouse lung fibroblasts (MLF), murine OSM (muOSM) stimulated high TIMP-1 mRNA expression in comparison to leukemia inhibitory factor (LIF), epidermal growth factor (EGF), interleukin (IL)-1beta and transforming growth factor (TGF)beta. In cell signaling, muOSM induced strong phosphorylation of extracellular-signal regulated protein kinase (Erk) 1/2, p38 and Akt in addition to phosphorylation of signal transducer and activator of transcription (STAT) 1, STAT3 and STAT5 within 15 min. LIF and TGFbeta had no such effects. EGF stimulated comparable or lower Erk1/2, p38 and Akt phosphorylation while IL-1beta induced p38 phosphorylation in the fibroblast cell lines. The Erk1/2 inhibitor PD98059 and the p38 inhibitor SB203580 inhibited TIMP-1 mRNA response to muOSM, whereas the phosphoinositide 3-kinase (PI3K) inhibitor LY294002 enhanced the TIMP-1 mRNA response in NIH 3T3 and MLg cells. PD98059 and SB203580, but not LY294002, also inhibited fold induction of a chloramphenicol acetyltransferase (CAT) reporter gene driven by a minimal TIMP-1 promoter that contained a proximal activator protein-1 (AP-1) site. Co-transfection with JunB or c-Jun expression vector in NIH 3T3 cells caused marked transactivation of the TIMP-1 promoter/CAT reporter gene. muOSM caused a rapid increase of JunB and c-Jun protein in NIH 3T3 cells. PD98059 partially inhibited the increase of JunB, but not c-Jun, whereas SB203580 did not induce detectable changes in expression of either AP-1 factor in response to muOSM. These results demonstrate that Erk1/2 and p38 contribute to the elevation of muOSM induced TIMP-1 expression, but PI3K does not, and suggest that Erk1/2 does so by enhancing JunB expression.
Collapse
Affiliation(s)
- Li Tong
- Department of Pathology and Molecular Medicine, Faculty of Health Sciences, McMaster University, HSC-4H17, 1200 Main Stree West, Hamilton, ON, Canada L8S 3Z5
| | | | | | | | | |
Collapse
|
48
|
Choi C, Jeong E, Benveniste EN. Caspase-1 mediates Fas-induced apoptosis and is up-regulated by interferon-gamma in human astrocytoma cells. J Neurooncol 2004; 67:167-76. [PMID: 15072464 DOI: 10.1023/b:neon.0000021896.52664.9e] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Resistance to Fas-mediated apoptosis contributes to tumor evasion from the host immune system and enables tumors to mediate alternative responses such as inflammation and angiogenesis. In this study, we investigated the molecular mechanisms of the resistance to Fas-mediated apoptosis and sensitization to Fas-induced cell death by IFN-gamma in human astrocytoma cells. To address this, we investigated the expression of thirty-three genes related to the Fas signal transduction pathways using RNase protection assay in five different human astrocytoma cells. Patterns of expression of these genes were similar between different cell lines and did not correlate with sensitivity to Fas-mediated cell death. Treatment with IFN-gamma increased the mRNA expression of caspases-1, -4 and -7 in addition to those of Fas and TRAIL in a time- and dose-dependent manner. Studies using specific caspase inhibitors showed that Fas-induced cell death was mediated by caspases-1, -3 and 8 in the Fas-sensitive human astrocytoma cell lines, CRT-J and U87-MG. We further demonstrated that these caspases were proteolytically cleaved upon Fas ligation in these cells. Interestingly, caspase-1 protein expression but not that of caspase-3 nor -8 was up-regulated by IFN-gamma only in Fas-sensitive CRT-J cells but not in Fas-resistant U373-MG cells. These results collectively suggest that caspase-1, along with caspases-3 and -8, mediate Fas-induced cell death in human astrocytoma cells, and post-transcriptional regulation of caspase-1 may determine the responsiveness to IFN-gamma-induced sensitization to Fas-mediated apoptosis.
Collapse
Affiliation(s)
- Chulhee Choi
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Seoul, Korea.
| | | | | |
Collapse
|
49
|
Isobe I, Maeno Y, Nagao M, Iwasa M, Koyama H, Seko-Nakamura Y, Monma-Ohtaki J. Cytoplasmic vacuolation in cultured rat astrocytes induced by an organophosphorus agent requires extracellular signal-regulated kinase activation. Toxicol Appl Pharmacol 2004; 193:383-92. [PMID: 14678747 DOI: 10.1016/j.taap.2003.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
There are various toxic chemicals that cause cell death. However, in certain cases deleterious agents elicit various cellular responses prior to cell death. To determine the cellular mechanisms by which such cellular responses are induced is important, but sufficient attention has not been paid to this issue to date. In this study, we showed the characteristic effects of an organophosphorus (OP) agent, bis(pinacolyl methyl)phosphonate (BPMP), which we synthesized for the study of OP nerve agents, on cultured rat astrocytes. Morphologically, BPMP induced cytoplasmic vacuolation and stellation in the rat astrocytes. Cytoplasmic vacuolation is a cell pathological change observed, for example, in vacuolar degeneration, and stellation has been reported in astrocytic reactions against various stimuli. By pretreatment with cycloheximide, a protein synthesis inhibitor, stellation was inhibited, although vacuolation was not. Cell staining with a mitochondrion-selective dye indicated that the vacuolation probably occurs in the mitochondria that are swollen and vacuolatred in the center. Interestingly, the extracellular signal-regulated kinase (ERK) cascade inhibitor inhibited vacuolation and, to some extent, stellation. These results suggest that the ERK signaling cascade is important for the induction of mitochondrial vacuolation. We expect that a detailed study of these astrocytic reactions will provide us new perspectives regarding the variation and pathological significance of cell morphological changes, such as vacuolar degeneration, and also the mechanisms underlying various neurological disorders.
Collapse
Affiliation(s)
- Ichiro Isobe
- Department of Forensic Medical Science, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan.
| | | | | | | | | | | | | |
Collapse
|
50
|
Repovic P, Fears CY, Gladson CL, Benveniste EN. Oncostatin-M induction of vascular endothelial growth factor expression in astroglioma cells. Oncogene 2003; 22:8117-24. [PMID: 14603252 DOI: 10.1038/sj.onc.1206922] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Oncostatin-M (OSM), a hematopoietic cytokine, and vascular endothelial growth factor (VEGF), a quintessential angiogenic signal, are coexpressed in development, cancer and inflammation. Here, we report that OSM treatment of human astroglioma cell lines increases VEGF levels by approximately threefold. Interleukin-1beta (IL-1beta), in combination with OSM, induces up to sevenfold higher VEGF expression, without significantly inducing VEGF on its own. Specifically examining the OSM contribution to VEGF expression, neutralizing antibodies to OSM receptor subunits gp130 and OSMRbeta, but not LIFRbeta, inhibited OSM induction of VEGF, indicating that the OSM-specific receptor OSMRbeta/gp130 transduces the OSM signal for VEGF synthesis. OSM induction of VEGF promoter activity maps to the (-1171, -786) region of the VEGF promoter, which contains a STAT-3-binding site. STAT-3 is indeed essential for this response, since overexpression of a dominant-negative STAT-3 blocks OSM induction of VEGF promoter activity, as well as endogenous VEGF expression. Finally, we demonstrate that OSM is expressed in glioblastoma multiforme tumor biopsies, a particularly malignant form of brain tumor. This novel mechanism of VEGF regulation in astroglioma cells may be active in pathophysiological states where both OSM and IL-1beta are present.
Collapse
Affiliation(s)
- Pavle Repovic
- Department of Cell Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | |
Collapse
|