1
|
Gaspa G, Cesarani A, Pauciullo A, Peana I, Macciotta NPP. Genomic Analysis of Sarda Sheep Raised at Diverse Temperatures Highlights Several Genes Involved in Adaptations to the Environment and Heat Stress Response. Animals (Basel) 2024; 14:3585. [PMID: 39765489 PMCID: PMC11672698 DOI: 10.3390/ani14243585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Livestock expresses complex traits influenced by several factors. The response of animals to variations in climatic factors, such as increases in temperature, may induce heat stress conditions. In this study, animals living at different temperatures were compared using the genome-wide Wright fixation index (FST). A total of 825 genotypes of Sarda breed ewes were divided into two groups based on the flocks' average temperature over a 20-year period to compute the FST: 395 and 430 sheep were represented in colder and hotter groups, respectively. After LOWESS regression and CONTROL CHART application, 623 significant markers and 97 selection signatures were found. A total of 280 positional candidate genes were retrieved from a public database. Among these genomic regions, we found 51 annotated genes previously associated with heat stress/tolerance in ruminants (FCGR1A, MDH1, UGP2, MYO1G, and HSPB3), as well as immune response and cellular mechanisms related to how animals cope with thermal stress (RIPK1, SERPINB1, SERPINB9, and PELI1). Moreover, other genes were associated with milk fat (SCD, HERC3, SCFD2, and CHUK), body weight, body fat, and intramuscular fat composition (AGPAT2, ABCD2, MFAP32, YTHDC1, SIRT3, SCD, and RNF121), which might suggest the influence of environmental conditions on the genome of Sarda sheep.
Collapse
Affiliation(s)
- Giustino Gaspa
- Department of Agricultural, Forest and Food Science, University of Torino, 10124 Torino, Italy;
| | - Alberto Cesarani
- Department of Agriculture, University of Sassari, 07100 Sassari, Italy; (A.C.); (N.P.P.M.)
- Department of Animal and Dairy Science, University of Georgia, Athens, GA 30602, USA
| | - Alfredo Pauciullo
- Department of Agricultural, Forest and Food Science, University of Torino, 10124 Torino, Italy;
| | - Ilaria Peana
- Servizio Agrometeorologico Regionale per la Sardegna (ARPAS), 07100 Sassari, Italy;
| | - Nicolò P. P. Macciotta
- Department of Agriculture, University of Sassari, 07100 Sassari, Italy; (A.C.); (N.P.P.M.)
| |
Collapse
|
2
|
Huang R, Zhang L, Li X, Liu F, Cheng X, Ran H, Wang Z, Li Y, Feng Y, Liang L, Su W, Melgiri ND, Sun Y. Anti-CXCR2 antibody-coated nanoparticles with an erythrocyte-platelet hybrid membrane layer for atherosclerosis therapy. J Control Release 2023; 356:610-622. [PMID: 36898531 DOI: 10.1016/j.jconrel.2023.02.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/20/2023] [Accepted: 02/26/2023] [Indexed: 03/12/2023]
Abstract
Atherosclerosis is the leading cause of mortality globally. RBC-platelet hybrid membrane-coated nanoparticles ([RBC-P]NPs), which biologically mimic platelets in vivo, display evidence of anti-atherosclerotic activity. The efficacy of a targeted RBC-platelet hybrid membrane-coated nanoparticles ([RBC-P]NP)-based approach was investigated as a primary preventive measure against atherosclerosis. A ligand-receptor interactome analysis conducted with circulating platelets and monocytes derived from CAD patients and healthy controls identified CXCL8-CXCR2 as a key platelet ligand-monocyte receptor dyad in CAD patients. Based on this analysis, a novel anti-CXCR2 [RBC-P]NP that specifically binds to CXCR2 and blocks the interaction between CXCL8 and CXCR2 was engineered and characterized. Administering anti-CXCR2 [RBC-P]NPs to Western diet-fed Ldlr-/- mice led to diminished plaque size, necrosis, and intraplaque macrophage accumulation relative to control [RBC-P]NPs or vehicle. Importantly, anti-CXCR2 [RBC-P]NPs demonstrated no adverse bleeding/hemorrhagic effects. A series of in vitro experiments was conducted to characterize anti-CXCR2 [RBC-P]NP's mechanism of action in plaque macrophages. Mechanistically, anti-CXCR2 [RBC-P]NPs inhibited p38α (Mapk14)-mediated, pro-inflammatory M1 skewing and corrected efferocytosis in plaque macrophages. This targeted [RBC-P]NP-based approach, in which the cardioprotective effects of anti-CXCR2 [RBC-P]NP therapy overweighs its bleeding/hemorrhagic risks, could potentially be used to proactively manage atherosclerotic progression in at-risk populations.
Collapse
Affiliation(s)
- Rongzhong Huang
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Municipality Clinical Research Center for Geriatrics and Gerontology, Chongqing 400010, China
| | - Lujun Zhang
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xingsheng Li
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Municipality Clinical Research Center for Geriatrics and Gerontology, Chongqing 400010, China
| | - Fan Liu
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Xiaoxiao Cheng
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Haitao Ran
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Zhigang Wang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China
| | - Yongyong Li
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Municipality Clinical Research Center for Geriatrics and Gerontology, Chongqing 400010, China
| | - Yuxing Feng
- Department of Rehabilitation and Pain Medicine, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Liwen Liang
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, China
| | - Wenhua Su
- Department of Cardiology, The First People's Hospital of Yunnan Province, Kunming, China
| | - N D Melgiri
- Impactys Foundation for Biomedical Research, San Diego, CA, USA
| | - Yang Sun
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ultrasound Molecular Imaging, Chongqing, China.
| |
Collapse
|
3
|
Zhang Z, Fan K, Meng J, Nie D, Zhao Z, Han Z. Deoxynivalenol hijacks the pathway of Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT-3) to drive caspase-3-mediated apoptosis in intestinal porcine epithelial cells. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 864:161058. [PMID: 36565876 DOI: 10.1016/j.scitotenv.2022.161058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/25/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
Deoxynivalenol (DON) can easily injure the intestinal tract, which represents the first barrier against food contaminants. The intestinal toxicity induced by DON was mainly focused on mitogen-activated protein kinase (MAPK) activation, however, the underlying mechanisms by which DON triggers apoptosis by other pathways remain poorly understood. In this study, the Janus kinase 2/signal transducers and activators of transcription 3 (JAK2/STAT-3) pathway was proposed to regulate the intrinsic apoptosis induced by DON and thoroughly investigated in intestinal porcine epithelial cells (IPEC-J2). First, DON was found to be able to efficiently inhibit cell viability and increase the release of lactate dehydrogenase. It could also enhance the activity of the cleaved caspase-3 in a time-dependent manner, accompanied by a loss of mitochondrial membrane potential and an up-regulation of the apoptosis rate. Then, the expression of genes associated with inflammation and apoptosis were investigated. DON increased the expression of IL-6, IL-1β, TNF-α, SOCS3 and Bax, but decreased the expression of Bcl-2 and Bcl-xL. Moreover, we discovered that DON robustly inhibited STAT-3 activity together with the down-regulation of JAK2, Bcl-2 and Bcl-xL, paralleling the increase in p38 phosphorylation. Furthermore, a pharmacological activation of JAK2/STAT-3 alleviated DON induced-apoptosis. Concurrent with the apoptotic pathway, during the initial exposure to DON (first 4 h), a survival pathway involving phosphorylated Erk1/2, Akt, and FoxO1 was also observed. Thus, apoptosis induced by DON was Janus faced: although the survival pathway was activated, the DON-induced apoptotic JAK2/STAT-3/caspase-3 pathway dominated, leading to an imbalance in cell homeostasis. This study provides a novel avenue to comprehensively reveal the pathological mechanisms of DON-induced intestinal disorders, which is promising for future applications to other contaminants in food and feed.
Collapse
Affiliation(s)
- Zhiqi Zhang
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Kai Fan
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Jiajia Meng
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Dongxia Nie
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Zhihui Zhao
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China
| | - Zheng Han
- Institute for Agro-food Standards and Testing Technology, Shanghai Academy of Agricultural Sciences, Shanghai 201403, China.
| |
Collapse
|
4
|
Yang X, Yan S, Wang P, Wang G. Identification of hub Genes in the Pathogenesis of Ischemic Stroke Based on Bioinformatics Analysis. J Korean Neurosurg Soc 2022; 65:697-709. [PMID: 35577542 PMCID: PMC9452394 DOI: 10.3340/jkns.2021.0200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 02/03/2022] [Indexed: 11/27/2022] Open
Abstract
Objective The present study aimed to identify the function of ischemic stroke (IS) patients’ peripheral blood and its role in IS, explore the pathogenesis, and provide direction for clinical research progress by comprehensive bioinformatics analysis.
Methods Two datasets, including GSE58294 and GSE22255, were downloaded from Gene Expression Omnibus database. GEO2R was utilized to obtain differentially expressed genes (DEGs). Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs were performed using the database annotation, visualization and integrated discovery database. The protein-protein interaction (PPI) network of DEGs was constructed by search tool of searching interactive gene and visualized by Cytoscape software, and then the Hub gene was identified by degree analysis. The microRNA (miRNA) and miRNA target genes closely related to the onset of stroke were obtained through the miRNA gene regulatory network.
Results In total, 36 DEGs, containing 27 up-regulated and nine down-regulated DEGs, were identified. GO functional analysis showed that these DEGs were involved in regulation of apoptotic process, cytoplasm, protein binding and other biological processes. KEGG enrichment analysis showed that these DEGs mediated signaling pathways, including HTLV-I infection and microRNAs in cancer. The results of PPI network and cytohubba showed that there was a relationship between DEGs, and five hub genes related to stroke were obtained : SOCS3, KRAS, PTGS2, EGR1, and DUSP1. Combined with the visualization of DEG-miRNAs, hsa-mir-16-5p, hsa-mir-181a-5p and hsa-mir-124-3p were predicted to be the key miRNAs in stroke, and three miRNAs were related to hub gene.
Conclusion Thirty-six DEGs, five Hub genes, and three miRNA were obtained from bioinformatics analysis of IS microarray data, which might provide potential targets for diagnosis and treatment of IS.
Collapse
Affiliation(s)
- Xitong Yang
- Genetic Testing Center, The First Affiliated hospital of Dali University, Dali 671000, Yunnan, China
| | - Shanquan Yan
- Clinical colllege of Dali University, Dali 671000, Yunnan, China
| | - Pengyu Wang
- Clinical colllege of Dali University, Dali 671000, Yunnan, China
| | - Guangming Wang
- Genetic Testing Center, The First Affiliated hospital of Dali University, Dali 671000, Yunnan, China
| |
Collapse
|
5
|
Sobah ML, Liongue C, Ward AC. SOCS Proteins in Immunity, Inflammatory Diseases, and Immune-Related Cancer. Front Med (Lausanne) 2021; 8:727987. [PMID: 34604264 PMCID: PMC8481645 DOI: 10.3389/fmed.2021.727987] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/16/2021] [Indexed: 01/10/2023] Open
Abstract
Cytokine signaling represents one of the cornerstones of the immune system, mediating the complex responses required to facilitate appropriate immune cell development and function that supports robust immunity. It is crucial that these signals be tightly regulated, with dysregulation underpinning immune defects, including excessive inflammation, as well as contributing to various immune-related malignancies. A specialized family of proteins called suppressors of cytokine signaling (SOCS) participate in negative feedback regulation of cytokine signaling, ensuring it is appropriately restrained. The eight SOCS proteins identified regulate cytokine and other signaling pathways in unique ways. SOCS1–3 and CISH are most closely involved in the regulation of immune-related signaling, influencing processes such polarization of lymphocytes and the activation of myeloid cells by controlling signaling downstream of essential cytokines such as IL-4, IL-6, and IFN-γ. SOCS protein perturbation disrupts these processes resulting in the development of inflammatory and autoimmune conditions as well as malignancies. As a consequence, SOCS proteins are garnering increased interest as a unique avenue to treat these disorders.
Collapse
Affiliation(s)
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Institue of Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, VIC, Australia.,Institue of Mental and Physical Health and Clinical Translation, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
6
|
Luo X, Chen XX, Qiao S, Li R, Lu Q, Geng R, Wang L, Zhou EM, Zhang G. Porcine reproductive and respiratory syndrome virus increases SOCS3 production via activation of p38/AP-1 signaling pathway to promote viral replication. Vet Microbiol 2021; 257:109075. [PMID: 33930700 DOI: 10.1016/j.vetmic.2021.109075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 04/18/2021] [Indexed: 12/15/2022]
Abstract
SOCS3 belongs to the suppressor of cytokine signaling (SOCS) family, which function as negative factors in host immune responses. Prior studies have noted the importance of SOCS family proteins in immunosuppression induced by some viruses. Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most important swine-borne viruses and has threatened the global swine industry with huge economic losses since it was first described in the 1980s. PRRSV is the etiological agent of PRRS, which causes reproductive failure and respiratory disorders. PRRSV causes immunosuppression thus establishing persistent infection. In this study, it was observed that SOCS3 was upregulated in PRRSV-infected primary porcine alveolar macrophages (PAMs) and Marc-145 cells with dose-dependent effects, which depends on virus replication. Deletion of AP-1 binding motif located in SOCS3 promoter inhibited promoter activities, which indicates that AP-1 is essential for PRRSV-induced SOCS3. This result was confirmed by experiments using AP-1 inhibitor, whose pretreatment suppressed SOCS3 mRNA and protein expression. Further research showed that p38 was crucial for PRRSV-induced SOCS3 production. Importantly, SOCS3 enhanced PRRSV replication during infection. Taken together, this study indicates that PRRSV infection induced SOCS3 expression through p38/AP-1 signaling pathway. These results revealed the molecular basis of SOCS3 upregulation and would advance further understanding of the strategy for viral immune evasion.
Collapse
Affiliation(s)
- Xuegang Luo
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China; College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Xin-Xin Chen
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Songlin Qiao
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Rui Li
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Qingxia Lu
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Rui Geng
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - Li Wang
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China
| | - En-Min Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Gaiping Zhang
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, 450002, China; College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China; College of Animal Medicine, Henan Agricultural University, Zhengzhou, 450002, China.
| |
Collapse
|
7
|
A data-driven computational model enables integrative and mechanistic characterization of dynamic macrophage polarization. iScience 2021; 24:102112. [PMID: 33659877 PMCID: PMC7895754 DOI: 10.1016/j.isci.2021.102112] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/01/2020] [Accepted: 01/21/2021] [Indexed: 01/09/2023] Open
Abstract
Macrophages are highly plastic immune cells that dynamically integrate microenvironmental signals to shape their own functional phenotypes, a process known as polarization. Here we develop a large-scale mechanistic computational model that for the first time enables a systems-level characterization, from quantitative, temporal, dose-dependent, and single-cell perspectives, of macrophage polarization driven by a complex multi-pathway signaling network. The model was extensively calibrated and validated against literature and focused on in-house experimental data. Using the model, we generated dynamic phenotype maps in response to numerous combinations of polarizing signals; we also probed into an in silico population of model-based macrophages to examine the impact of polarization continuum at the single-cell level. Additionally, we analyzed the model under an in vitro condition of peripheral arterial disease to evaluate strategies that can potentially induce therapeutic macrophage repolarization. Our model is a key step toward the future development of a network-centric, comprehensive "virtual macrophage" simulation platform.
Collapse
|
8
|
Bongartz H, Seiß EA, Bock J, Schaper F. Glucocorticoids attenuate interleukin-6-induced c-Fos and Egr1 expression and impair neuritogenesis in PC12 cells. J Neurochem 2021; 157:532-549. [PMID: 33454999 DOI: 10.1111/jnc.15305] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/07/2021] [Accepted: 01/10/2021] [Indexed: 01/15/2023]
Abstract
Interleukin-6 (IL-6) is a cytokine primarily known for immune regulation. There is also growing evidence that IL-6 triggers neurogenesis and impacts neural development, both life-long occurring processes that can be impaired by early-life and adult stress. Stress induces the release of glucocorticoids by activation of the hypothalamic-pituitary-adrenal (HPA) axis. On the cellular level, glucocorticoids act via the ubiquitously expressed glucocorticoid receptor. Thus, we aimed to elucidate whether glucocorticoids affect IL-6-induced neural development. Here, we show that IL-6 signalling induces neurite outgrowth in adrenal pheochromocytoma PC12 cells in a mitogen-activated protein kinase (MAPK) pathway-dependent manner, since neurite outgrowth was diminished upon Mek-inhibitor treatment. Using quantitative biochemical approaches, such as qRT-PCR analysis of Hyper-IL-6 treated PC12 cells, we show that neurite outgrowth induced by IL-6 signalling is accompanied by early and transient MAPK-dependent mRNA expression of immediate early genes coding for proteins such as early growth response protein 1 (Egr1) and c-Fos. This correlates with reduced proliferation and prolonged G0/G1 cell cycle arrest as determined by monitoring the cellular DNA content using flow cytometry. These results indicate for IL-6 signalling-induced neural differentiation. Interestingly, the glucocorticoid Dexamethasone impairs early IL-6 signalling-induced mRNA expression of c-Fos and Egr1 and restrains neurite outgrowth. Impaired Egr1 and c-Fos expression in neural development is implicated in the aetiology of neuropathologies. Thus, it appears likely that stress-induced release of glucocorticoids, as well as therapeutically administered glucocorticoids, contribute to the development of neuropathologies by reducing the expression of Egr1 and c-Fos, and by restraining IL-6-dependent neural differentiation.
Collapse
Affiliation(s)
- Hannes Bongartz
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Magdeburg, Germany
| | - Elena Anne Seiß
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Magdeburg, Germany
| | - Jörg Bock
- Institute of Biology, PG "Epigenetics and Structural Plasticity", Otto-von-Guericke University, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Otto-von-Guericke University, Magdeburg, Germany
| | - Fred Schaper
- Institute of Biology, Department of Systems Biology, Otto-von-Guericke University, Magdeburg, Germany.,Center for Dynamic Systems: Systems Engineering (CDS), Otto-von-Guericke University, Magdeburg, Germany.,Magdeburg Center for Systems Biology (MACS), Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
9
|
Hajj GNM, Nunes PBC, Roffe M. Genome-wide translation patterns in gliomas: An integrative view. Cell Signal 2020; 79:109883. [PMID: 33321181 DOI: 10.1016/j.cellsig.2020.109883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Gliomas are the most frequent tumors of the central nervous system (CNS) and include the highly malignant glioblastoma (GBM). Characteristically, gliomas have translational control deregulation related to overactivation of signaling pathways such as PI3K/AKT/mTORC1 and Ras/ERK1/2. Thus, mRNA translation appears to play a dominant role in glioma gene expression patterns. The, analysis of genome-wide translated transcripts, together known as the translatome, may reveal important information for understanding gene expression patterns in gliomas. This review provides a brief overview of translational control mechanisms altered in gliomas with a focus on the current knowledge related to the translatomes of glioma cells and murine glioma models. We present an integrative meta-analysis of selected glioma translatome data with the aim of identifying recurrent patterns of gene expression preferentially regulated at the level of translation and obtaining clues regarding the pathological significance of these alterations. Re-analysis of several translatome datasets was performed to compare the translatomes of glioma models with those of their non-tumor counterparts and to document glioma cell responses to radiotherapy and MNK modulation. The role of recurrently altered genes in the context of translational control and tumorigenesis are discussed.
Collapse
Affiliation(s)
- Glaucia Noeli Maroso Hajj
- International Research Institute, A.C.Camargo Cancer Center, Rua Taguá, 440, São Paulo ZIP Code: 01508-010, Brazil; National Institute of Oncogenomics and Innovation, Brazil.
| | - Paula Borzino Cordeiro Nunes
- International Research Institute, A.C.Camargo Cancer Center, Rua Taguá, 440, São Paulo ZIP Code: 01508-010, Brazil
| | - Martin Roffe
- International Research Institute, A.C.Camargo Cancer Center, Rua Taguá, 440, São Paulo ZIP Code: 01508-010, Brazil; National Institute of Oncogenomics and Innovation, Brazil.
| |
Collapse
|
10
|
Brouwer MAE, Jones-Warner W, Rahman S, Kerstholt M, Ferreira AV, Oosting M, Hooiveld GJ, Netea MG, Joosten LAB. B. burgdorferi sensu lato-induced inhibition of antigen presentation is mediated by RIP1 signaling resulting in impaired functional T cell responses towards Candida albicans. Ticks Tick Borne Dis 2020; 12:101611. [PMID: 33360386 DOI: 10.1016/j.ttbdis.2020.101611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/22/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023]
Abstract
Antigen presentation is a crucial innate immune cell function that instructs adaptive immune cells. Loss of this pathway severely impairs the development of adaptive immune responses. To investigate whether B. burgdorferi sensu lato. spirochetes modulate the induction of an effective immune response, primary human PBMCs were isolated from healthy volunteers and stimulated with B. burgdorferi s.l. Through cell entry, TNF receptor I, and RIP1 signaling cascades, B. burgdorferi s.l. strongly downregulated genes and proteins involved in antigen presentation, specifically HLA-DM, MHC class II and CD74. Antigen presentation proteins were distinctively inhibited in monocyte subsets, monocyte-derived macrophages, and dendritic cells. When compared to a range of other pathogens, B. burgdorferi s.l.-induced suppression of antigen presentation appears to be specific. Inhibition of antigen presentation interfered with T-cell recognition of B. burgdorferi s.l., and memory T-cell responses against Candidaalbicans. Re-stimulation of PBMCs with the commensal microbe C.albicans following B. burgdorferi s.l. exposure resulted in significantly reduced IFN-γ, IL-17 and IL-22 production. These findings may explain why patients with Lyme borreliosis develop delayed adaptive immune responses. Unravelling the mechanism of B. burgdorferi s.l.-induced inhibition of antigen presentation, via cell entry, TNF receptor I, and RIP1 signaling cascades, explains the difficulty to diagnose the disease based on serology and to obtain an effective vaccine against Lyme borreliosis.
Collapse
Affiliation(s)
- Michelle A E Brouwer
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - William Jones-Warner
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Shafaque Rahman
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Mariska Kerstholt
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Anaísa V Ferreira
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Marije Oosting
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands
| | - Guido J Hooiveld
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands; Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Center for Infectious Diseases (RCI), Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
11
|
Sarajlic M, Neuper T, Vetter J, Schaller S, Klicznik MM, Gratz IK, Wessler S, Posselt G, Horejs-Hoeck J. H. pylori modulates DC functions via T4SS/TNFα/p38-dependent SOCS3 expression. Cell Commun Signal 2020; 18:160. [PMID: 33023610 PMCID: PMC7541176 DOI: 10.1186/s12964-020-00655-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 09/06/2020] [Indexed: 12/11/2022] Open
Abstract
Background Helicobacter pylori (H. pylori) is a gram-negative bacterium that chronically infects approximately 50% of the world’s human population. While in most cases the infection remains asymptomatic, 10% of infected individuals develop gastric pathologies and 1–3% progress to gastric cancer. Although H. pylori induces severe inflammatory responses, the host’s immune system fails to clear the pathogen and H. pylori can persist in the human stomach for decades. As suppressor of cytokine signaling (SOCS) proteins are important feedback regulators limiting inflammatory responses, we hypothesized that H. pylori could modulate the host’s immune responses by inducing SOCS expression. Methods The phenotype of human monocyte-derived DCs (moDCs) infected with H. pylori was analyzed by flow cytometry and multiplex technology. SOCS expression levels were monitored by qPCR and signaling studies were conducted by means of Western blot. For functional studies, RNA interference-based silencing of SOCS1–3 and co-cultures with CD4+ T cells were performed. Results We show that H. pylori positive gastritis patients express significantly higher SOCS3, but not SOCS1 and SOCS2, levels compared to H. pylori negative patients. Moreover, infection of human moDCs with H. pylori rapidly induces SOCS3 expression, which requires the type IV secretion system (T4SS), release of TNFα, and signaling via the MAP kinase p38, but appears to be independent of TLR2, TLR4, MEK1/2 and STAT proteins. Silencing of SOCS3 expression in moDCs prior to H. pylori infection resulted in increased release of both pro- and anti-inflammatory cytokines, upregulation of PD-L1, and decreased T-cell proliferation. Conclusions This study shows that H. pylori induces SOCS3 via an autocrine loop involving the T4SS and TNFα and p38 signaling. Moreover, we demonstrate that high levels of SOCS3 in DCs dampen PD-L1 expression on DCs, which in turn drives T-cell proliferation. Video Abstract
Collapse
Affiliation(s)
- Muamera Sarajlic
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Theresa Neuper
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Julia Vetter
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Hagenberg im Muehlkreis, Austria
| | - Susanne Schaller
- Bioinformatics Research Group, University of Applied Sciences Upper Austria, Hagenberg im Muehlkreis, Austria
| | - Maria M Klicznik
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Iris K Gratz
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Silja Wessler
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Gernot Posselt
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria
| | - Jutta Horejs-Hoeck
- Department of Biosciences, University of Salzburg, Hellbrunner Strasse 34, 5020, Salzburg, Austria.
| |
Collapse
|
12
|
Guo J, Mo J, Zhao Q, Han Q, Kanerva M, Iwata H, Li Q. De novo transcriptomic analysis predicts the effects of phenolic compounds in Ba River on the liver of female sharpbelly (Hemiculter lucidus). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 264:114642. [PMID: 32408079 DOI: 10.1016/j.envpol.2020.114642] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/18/2020] [Accepted: 04/19/2020] [Indexed: 06/11/2023]
Abstract
This work aimed at predicting the toxic effects of phenolic compounds in Ba River on the health of female sharpbelly (Hemiculter lucidus) by the de novo transcriptomic analysis of the liver. Sharpbelly, a native fish living in freshwater ecosystem of East Asia, were sampled upstream, near, and downstream of a wastewater discharge to the Ba river. Based on the occurrence of bisphenol A (BPA), nonylphenol (NP), and 4-tert-octylphenol (4-t-OP) in the water and fish sampled from each site, up-, mid-, and down-stream were interpreted as control, high, and low treatment groups, respectively. In the mid-stream group the Fulton's condition factor (CF) and body weight were remarkably increased by approximate 20%; the gonado-somatic index (GSI) and hepatosomatic index (HSI) in mid-stream fish showed a similar increasing trend but lacking of statistical difference. Exposure to wastewater effluent caused 160 and 162 differentially expressed genes (DEGs) in up-mid and down-mid stream groups, respectively. Two sets of DEGs were primarily enriched in the signaling pathways of drug metabolism, endocrine system, cellular process, and lipid metabolism in the mid-stream sharpbelly, which may alter the fish behavior, disrupt the reproductive function, and lead to hypothyroidism, hepatic steatosis, etc. Taken together, our results linked the disrupted signaling pathways with activities of phenolic compounds to predict the potential effects of wastewater effluent on the health of wild fish.
Collapse
Affiliation(s)
- Jiahua Guo
- Shaanxi Key Laboratory of Earth Surface System and Environmental Carrying Capacity, College of Urban and Environmental Sciences, Northwest University, Xi'an, 710127, China
| | - Jiezhang Mo
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Qian Zhao
- Shaanxi Key Laboratory of Earth Surface System and Environmental Carrying Capacity, College of Urban and Environmental Sciences, Northwest University, Xi'an, 710127, China
| | - Qizhi Han
- Shaanxi Key Laboratory of Earth Surface System and Environmental Carrying Capacity, College of Urban and Environmental Sciences, Northwest University, Xi'an, 710127, China
| | - Mirella Kanerva
- Center for Marine Environmental Studies, Ehime University, Bunkyo-cho 2-5, Matsuyama, Ehime prefecture, 790-8577, Japan
| | - Hisato Iwata
- Center for Marine Environmental Studies, Ehime University, Bunkyo-cho 2-5, Matsuyama, Ehime prefecture, 790-8577, Japan
| | - Qi Li
- Shaanxi Key Laboratory of Earth Surface System and Environmental Carrying Capacity, College of Urban and Environmental Sciences, Northwest University, Xi'an, 710127, China.
| |
Collapse
|
13
|
Aziz SGG, Aziz SGG, Khabbazi A, Alipour S. The methylation status of TNF-α and SOCS3 promoters and the regulation of these gene expressions in patients with Behçet's disease. Biomarkers 2020; 25:384-390. [PMID: 32475174 DOI: 10.1080/1354750x.2020.1754912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction: The aim of this study was to evaluate the methylation status of TNF-α and SOCS3 promoters in patients with BD and compare them with a healthy group.Method: This was a case-control study, in which 47 subjects with BD and 61 individuals as the control participated. Blood samples were collected from all the participants. Then, PBMCs were isolated using the Ficoll method and methylation of considered sites was investigated using the qMS-PCR technique after DNA extraction by the rapid genomic DNA extraction method and its analysis with Nano-drop.Results: The methylation and expression of TNF-α showed that the methylation level significantly declined in the patient in comparison with the healthy (p < 0.05). Moreover, the results on the mean expression showed that it significantly increased in the patient group, as compared with the healthy group (p < 0.05). In addition, the expression of the SOCS3 gene was not significantly different between the patients and healthy subjects while the level of SOCS3 methylation was significantly higher in the patient group than that in the healthy group (p < 0.05).Discussion: The present study revealed that the gene expression of TNF-alpha increased in BD patients, suggesting that TNF-alpha likely has a role in the pathogenesis of BD.
Collapse
Affiliation(s)
| | - Sara Gholizadeh-Ghaleh Aziz
- Department of Food Science and Technology, College of Agriculture, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Alireza Khabbazi
- Connective Tissue Disease, Tabriz University of Medical Science, Tabriz, Iran
| | - Shahriar Alipour
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
14
|
Rancourt RC, Ott R, Ziska T, Schellong K, Melchior K, Henrich W, Plagemann A. Visceral Adipose Tissue Inflammatory Factors (TNF-Alpha, SOCS3) in Gestational Diabetes (GDM): Epigenetics as a Clue in GDM Pathophysiology. Int J Mol Sci 2020; 21:ijms21020479. [PMID: 31940889 PMCID: PMC7014132 DOI: 10.3390/ijms21020479] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/19/2022] Open
Abstract
Gestational diabetes (GDM) is among the most challenging diseases in westernized countries, affecting mother and child, immediately and in later life. Obesity is a major risk factor for GDM. However, the impact visceral obesity and related epigenetics play for GDM etiopathogenesis have hardly been considered so far. Our recent findings within the prospective ‘EaCH’ cohort study of women with GDM or normal glucose tolerance (NGT), showed the role, critical factors of insulin resistance (i.e., adiponectin, insulin receptor) may have for GDM pathophysiology with epigenetically modified expression in subcutaneous (SAT) and visceral (VAT) adipose tissues. Here we investigated the expression and promoter methylation of key inflammatory candidates, tumor necrosis factor-alpha (TNF-α) and suppressor of cytokine signaling 3 (SOCS3) in maternal adipose tissues collected during caesarian section (GDM, n = 19; NGT, n = 22). The mRNA expression of TNF-α and SOCS3 was significantly increased in VAT, but not in SAT, of GDM patients vs. NGT, accompanied by specific alterations of respective promoter methylation patterns. In conclusion, we propose a critical role of VAT and visceral obesity for the pathogenesis of GDM, with epigenetic alterations of the expression of inflammatory factors as a potential factor.
Collapse
Affiliation(s)
- Rebecca C. Rancourt
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (R.O.); (T.Z.); (K.S.); (K.M.); (A.P.)
- Correspondence:
| | - Raffael Ott
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (R.O.); (T.Z.); (K.S.); (K.M.); (A.P.)
| | - Thomas Ziska
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (R.O.); (T.Z.); (K.S.); (K.M.); (A.P.)
| | - Karen Schellong
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (R.O.); (T.Z.); (K.S.); (K.M.); (A.P.)
| | - Kerstin Melchior
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (R.O.); (T.Z.); (K.S.); (K.M.); (A.P.)
| | - Wolfgang Henrich
- Clinic of Obstetrics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany;
| | - Andreas Plagemann
- Division of ‘Experimental Obstetrics’, Clinic of Obstetrics, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow-Klinikum, 13353 Berlin, Germany; (R.O.); (T.Z.); (K.S.); (K.M.); (A.P.)
| |
Collapse
|
15
|
Jiang X, He M, Bai J, Chan CB, Wong AOL. Signal Transduction for TNFα-Induced Type II SOCS Expression and Its Functional Implication in Growth Hormone Resistance in Carp Hepatocytes. Front Endocrinol (Lausanne) 2020; 11:20. [PMID: 32082258 PMCID: PMC7003395 DOI: 10.3389/fendo.2020.00020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 01/13/2020] [Indexed: 01/15/2023] Open
Abstract
In mammals, local production of tumor necrosis factor α (TNFα) inhibits growth hormone (GH)-induced IGF-I expression at tissue level and contributes to GH resistance caused by sepsis/endotoxemia and inflammation. Although the loss of GH responsiveness can be mediated by a parallel rise in SOCS expression, the signaling mechanisms for TNFα-induced SOCS expression at the hepatic level have not been characterized and the comparative aspects of the phenomenon, especially in lower vertebrates, are still unknown. Recently, type II SOCS, including SOCS1-3 and CISH, have been cloned in grass carp and shown to act as the feedback repressors for GH signaling via JAK2/STAT5 pathway. To shed light on the mechanisms for TNFα-induced GH resistance in fish model, grass carp TNFα was cloned and confirmed to be a single-copy gene expressed in various tissues including the liver. In carp hepatocytes, incubation with the endotoxin LPS induced TNFα expression with parallel rises in SOCS1-3 and CISH mRNA levels. Similar to LPS, TNFα treatment could block GH-induced IGF-I/-II mRNA expression and elevate SOCS1, SOCS3, and CISH transcript levels. However, TNFα was not effective in altering SOCS2 expression. In parallel experiment, LPS blockade of IGF-I/-II signals caused by GH could be partially reverted by TNFα receptor antagonism. At hepatocyte level, TNFα induction also triggered rapid phosphorylation of IκBα, MEK1/2, ERK1/2, MKK3/6, P38MAPK, Akt, JAK2, and STAT1,3,5, and TNFα-induced SOCS1, SOCS3, and CISH mRNA expression could be negated by inhibiting the IKK/NFκB, MAPK, PI3K/Akt, and JAK/STAT cascades. Our findings, as a whole, suggest that local production of TNFα may interfere with IGF-I/-II induction by GH in the carp liver by up-regulation of SOCS1, SOCS3, and CISH via IKK/NFκB, MAPK, PI3K/Akt, and JAK/STAT-dependent mechanisms, which may contribute to GH resistance induced by endotoxin in carp species.
Collapse
|
16
|
Luo X, Chen XX, Qiao S, Li R, Xie S, Zhou X, Deng R, Zhou EM, Zhang G. Porcine Reproductive and Respiratory Syndrome Virus Enhances Self-Replication via AP-1-Dependent Induction of SOCS1. THE JOURNAL OF IMMUNOLOGY 2019; 204:394-407. [PMID: 31826939 DOI: 10.4049/jimmunol.1900731] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/07/2019] [Indexed: 12/25/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has caused tremendous economic losses in the swine industry since its emergence in the late 1980s. PRRSV exploits various strategies to evade immune responses and establish chronic persistent infections. Suppressor of cytokine signaling (SOCS) 1, a member of the SOCS family, is a crucial intracellular negative regulator of innate immunity. In this study, it was shown that SOCS1 can be co-opted by PRRSV to evade host immune responses, facilitating viral replication. It was observed that PRRSV induced SOCS1 production in porcine alveolar macrophages, monkey-derived Marc-145 cells, and porcine-derived CRL2843-CD163 cells. SOCS1 inhibited the expression of IFN-β and IFN-stimulated genes, thereby markedly enhancing PRRSV replication. It was observed that the PRRSV N protein has the ability to upregulate SOCS1 production and that nuclear localization signal-2 (NLS-2) is essential for SOCS1 induction. Moreover, SOCS1 upregulation was dependent on p38/AP-1 and JNK/AP-1 signaling pathways rather than classical type I IFN signaling pathways. In summary, to our knowledge, the findings of this study uncovered the molecular mechanism that underlay SOCS1 induction during PRRSV infection, providing new insights into viral immune evasion and persistent infection.
Collapse
Affiliation(s)
- Xuegang Luo
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, People's Republic of China.,Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People's Republic of China; and
| | - Xin-Xin Chen
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People's Republic of China; and
| | - Songlin Qiao
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People's Republic of China; and
| | - Rui Li
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People's Republic of China; and
| | - Sha Xie
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People's Republic of China; and
| | - Xinyu Zhou
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People's Republic of China; and
| | - Ruiguang Deng
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People's Republic of China; and
| | - En-Min Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, People's Republic of China
| | - Gaiping Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, People's Republic of China; .,Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou 450002, People's Republic of China; and.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, Jiangsu, People's Republic of China
| |
Collapse
|
17
|
Klepsch O, Namer LS, Köhler N, Kaempfer R, Dittrich A, Schaper F. Intragenic regulation of SOCS3 isoforms. Cell Commun Signal 2019; 17:70. [PMID: 31238931 PMCID: PMC6593527 DOI: 10.1186/s12964-019-0379-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/01/2019] [Indexed: 11/13/2022] Open
Abstract
Background Inflammatory reactions are commonly affected by stress responses. Interleukin-6 signalling is part of the inflammatory response and is stringently regulated by the feedback inhibitor SOCS3 expressed in a short and long isoform. Here, we studied the inhibitory potential of the two SOCS3 isoforms. Furthermore, we analysed the regulation of SOCS3 isoform expression and the role of PKR stress kinase signalling in SOCS3 protein expression. Methods We performed Western blotting, reporter assays, genetic analyses and manipulations for studying SOCS3 isoform expression and activation of signalling components involved in interleukin-6-induced and PKR-dependent signalling. Results Interleukin-6-induced endogenous expression of both SOCS3 isoforms was found in distinct cell types. Forced expression of either the long or short SOCS3 isoform demonstrated equal inhibitory activity of each isoform and confirmed longer half-life of the short isoform. Study of intragenic regulation of SOCS3 isoform expression revealed that (i) the 5′-UTR of SOCS3 mRNA restrains specifically expression of the long SOCS3 isoform, (ii) expression of the long isoform restrains expression of the short isoform, and (iii) signalling through the stress kinase PKR does not impact on SOCS3 isoform ratio. Conclusions Both SOCS3 isoforms show a similar potential for inhibiting interleukin-6 signalling but differ in their half-lives. Relative expression of the isoforms depends on intragenic elements yet is independent of PKR signalling. Graphic abstract ![]()
Electronic supplementary material The online version of this article (10.1186/s12964-019-0379-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Oliver Klepsch
- Department of Systems Biology, Institute of Biology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Lise Sarah Namer
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Nadine Köhler
- Department of Systems Biology, Institute of Biology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - Raymond Kaempfer
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, 9112102, Jerusalem, Israel
| | - Anna Dittrich
- Department of Systems Biology, Institute of Biology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany.
| | - Fred Schaper
- Department of Systems Biology, Institute of Biology, Otto-von-Guericke University Magdeburg, Universitätsplatz 2, 39106, Magdeburg, Germany.
| |
Collapse
|
18
|
Convery O, Gargan S, Kickham M, Schroder M, O'Farrelly C, Stevenson NJ. The hepatitis C virus (HCV) protein, p7, suppresses inflammatory responses to tumor necrosis factor (TNF)-α via signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK)-mediated induction of suppressor of cytokine signaling (SOCS)3. FASEB J 2019; 33:8732-8744. [PMID: 31163989 DOI: 10.1096/fj.201800629rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Viruses use a spectrum of immune evasion strategies that enable infection and replication. The acute phase of hepatitis C virus (HCV) infection is characterized by nonspecific and often mild clinical symptoms, suggesting an immunosuppressive mechanism that, unless symptomatic liver disease presents, allows the virus to remain largely undetected. We previously reported that HCV induced the regulatory protein suppressor of cytokine signaling (SOCS)3, which inhibited TNF-α-mediated inflammatory responses. However, the mechanism by which HCV up-regulates SOCS3 remains unknown. Here we show that the HCV protein, p7, enhances both SOCS3 mRNA and protein expression. A p7 inhibitor reduced SOCS3 induction, indicating that p7's ion channel activity was required for optimal up-regulation of SOCS3. Short hairpin RNA and chemical inhibition revealed that both the Janus kinase-signal transducer and activator of transcription (JAK-STAT) and MAPK pathways were required for p7-mediated induction of SOCS3. HCV-p7 expression suppressed TNF-α-mediated IκB-α degradation and subsequent NF-κB promoter activity, revealing a new and functional, anti-inflammatory effect of p7. Together, these findings identify a molecular mechanism by which HCV-p7 induces SOCS3 through STAT3 and ERK activation and demonstrate that p7 suppresses proinflammatory responses to TNF-α, possibly explaining the lack of inflammatory symptoms observed during early HCV infection.-Convery, O., Gargan, S., Kickham, M., Schroder, M., O'Farrelly, C., Stevenson, N. J. The hepatitis C virus (HCV) protein, p7, suppresses inflammatory responses to tumor necrosis factor (TNF)-α via signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK)-mediated induction of suppressor of cytokine signaling (SOCS)3.
Collapse
Affiliation(s)
- Orla Convery
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Siobhan Gargan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | | | | | - Cliona O'Farrelly
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.,School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Nigel J Stevenson
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
19
|
DiNicolantonio JJ, McCarty M, OKeefe J. Does elevated bilirubin aid weight control by preventing development of hypothalamic leptin resistance? Open Heart 2019; 6:e000897. [PMID: 30997121 PMCID: PMC6443125 DOI: 10.1136/openhrt-2018-000897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/04/2019] [Indexed: 12/16/2022] Open
Affiliation(s)
| | | | - James OKeefe
- Preventive Cardiology, Mid America Heart Institute, Kansas City, Missouri, USA
| |
Collapse
|
20
|
Abstract
Iron, an essential nutrient, is required for many biological processes but is also toxic in excess. The lack of a mechanism to excrete excess iron makes it crucial for the body to regulate the amount of iron absorbed from the diet. This regulation is mediated by the hepatic hormone hepcidin. Hepcidin also controls iron release from macrophages that recycle iron and from hepatocytes that store iron. Hepcidin binds to the only known iron export protein, ferroportin, inducing its internalization and degradation and thus limiting the amount of iron released into the plasma. Important regulators of hepcidin, and therefore of systemic iron homeostasis, include plasma iron concentrations, body iron stores, infection and inflammation, hypoxia and erythropoiesis, and, to a lesser extent, testosterone. Dysregulation of hepcidin production contributes to the pathogenesis of many iron disorders: hepcidin deficiency causes iron overload in hereditary hemochromatosis and non-transfused β-thalassemia, whereas overproduction of hepcidin is associated with iron-restricted anemias seen in patients with chronic inflammatory diseases and inherited iron-refractory iron-deficiency anemia. The present review summarizes our current understanding of the molecular mechanisms and signaling pathways contributing to hepcidin regulation by these factors and highlights the issues that still need clarification.
Collapse
Affiliation(s)
- Marie-Paule Roth
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.
| | - Delphine Meynard
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Hélène Coppin
- Institut de Recherche en Santé Digestive (IRSD), Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| |
Collapse
|
21
|
HuoXueJieDu Formula Alleviates Diabetic Retinopathy in Rats by Inhibiting SOCS3-STAT3 and TIMP1-A2M Pathways. Int J Genomics 2017; 2017:4832125. [PMID: 29318137 PMCID: PMC5727685 DOI: 10.1155/2017/4832125] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/30/2017] [Accepted: 10/08/2017] [Indexed: 02/07/2023] Open
Abstract
HuoXueJieDu (HXJD) formula exerts protective effects against diabetic retinopathy (DR) in rats, but its underlying mechanism remains unknown. In the present study, the diabetic rats were established using streptozocin. The administration of HXJD was initiated at 20 weeks after diabetes induction and continued for 12 weeks. Whole genome expression profiles in rat retinas were examined using microarray technology. Differential gene expression and pathway enrichment analysis were conducted on the microarray data, with validation through real-time PCR and immunohistochemical staining. The results showed that 170 genes and several IPA canonical pathways related to inflammation, matrix metabolism, and phototransduction were regulated by HXJD. PCR validation of selected genes, including SOCS3, STAT3, TIMP1, and A2M, confirmed the gene expression changes influenced by HXJD. In addition, the immunohistochemical staining results suggested that critical members of the SOCS3-STAT3 pathway were also affected by HXJD. Taken together, these results indicated that SOCS3-STAT3 and TIMP1-A2M pathways might mediate the alleviation of HXJD activities in rats with diabetic retinopathy.
Collapse
|
22
|
Xiong H, Zhang Y, Chen S, Ni Z, He J, Li X, Li B, Zhao K, Yang F, Zeng Y, Chen B, He F. Induction of SOCS3 by liver X receptor suppresses the proliferation of hepatocellular carcinoma cells. Oncotarget 2017; 8:64083-64094. [PMID: 28969053 PMCID: PMC5609985 DOI: 10.18632/oncotarget.19321] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/10/2017] [Indexed: 02/06/2023] Open
Abstract
Liver X receptor (LXR), a member of nuclear receptor superfamily, is involved in the regulation of glucose, lipid and cholesterol metabolism. Recently, it has been reported that LXR suppress different kinds of cancers including hepatocellular carcinoma (HCC). However, the corresponding mechanism is still not well elucidated. In the present study, we found that activation of LXR downregulated cyclin D1 while upregulated p21 and p27 by elevating the level of suppressor of cytokine signaling 3 (SOCS3), leading to the cell cycle arrest at G1/S phase and growth inhibition of HCC cells. Moreover, we demonstrated that LXRα (not LXRβ) mediated the induction of SOCS3 in HCC cells. Subsequently, we showed that LXR activation enhanced the mRNA stability of SOCS3, but had no significant influence on the transcriptional activity of SOCS3 gene promoter. The experiments in nude mice revealed that LXR agonist inhibited the growth of xenograft tumors and enhanced SOCS3 expression in vivo. These results indicate that “LXRα-SOCS3-cyclin D1/p21/p27” is a novel pathway by which LXR exerts its anti-HCC effects, suggesting that the pathway may be a new potential therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Haojun Xiong
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Yan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Shan Chen
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Zhenhong Ni
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Jintao He
- Battalion 17 of Students, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Xinzhe Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Bo Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Kai Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Fan Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Yijun Zeng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Bingbo Chen
- Laboratory Animal Center, Third Military Medical University, Chongqing 400038, China
| | - Fengtian He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
23
|
Iwahara N, Hisahara S, Kawamata J, Matsumura A, Yokokawa K, Saito T, Fujikura M, Manabe T, Suzuki H, Matsushita T, Suzuki S, Shimohama S. Role of Suppressor of Cytokine Signaling 3 (SOCS3) in Altering Activated Microglia Phenotype in APPswe/PS1dE9 Mice. J Alzheimers Dis 2017; 55:1235-1247. [PMID: 27814300 DOI: 10.3233/jad-160887] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In response to changes of the central nervous system environment, microglia are capable of acquiring diverse phenotypes for cytotoxic or immune regulation and resolution of injury. Alzheimer's disease (AD) pathology also induces several microglial activations, resulting in production of pro-inflammatory cytokines and reactive oxygen species or clearance of amyloid-β (Aβ) through phagocytosis. We previously demonstrated that microglial activation and increase in oxidative stress started from the middle age in APPswe/PS1dE9 mice, and hypothesized that M1 activation occurs in middle-aged AD mice by Aβ stimulation. In the present study, we analyzed in vivo expressions of pro-inflammatory cytokines (M1 microglial markers), M2 microglial markers, and suppressor of cytokine signaling (SOCS) family, and examined the microglial phenotypic profile in APPswe/PS1dE9 mice. Then we compared the in vitro gene expression patterns of Aβ- and lipopolysaccharide (LPS)-stimulated primary-cultured microglia. Microglia in APPswe/PS1dE9 mice exhibited an M1-like phenotype, expressing tumor necrosis factor α (TNFα) but not interleukin 6 (IL6). Aβ-stimulated primary-cultured microglia also expressed TNFα but not IL6, whereas LPS-stimulated primary-cultured microglia expressed both pro-inflammatory cytokines. Furthermore, both microglia in APPswe/PS1dE9 mice and Aβ-stimulated primary-cultured microglia expressed SOCS3. Reduction of SOCS3 expression in Aβ-challenged primary-cultured microglia resulted in upregulation of IL6 expression. Our findings indicate that SOCS3 suppresses complete polarization to M1 phenotype through blocking IL6 production, and Aβ-challenged primary-cultured microglia replicate the in vivo gene expression pattern of microglia in APPswe/PS1dE9 mice. Aβ may induce the M1-like phenotype through blocking of IL6 by SOCS3.
Collapse
Affiliation(s)
- Naotoshi Iwahara
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Shin Hisahara
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Jun Kawamata
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Akihiro Matsumura
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Kazuki Yokokawa
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Taro Saito
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Mai Fujikura
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Tatsuo Manabe
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Hiromi Suzuki
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Takashi Matsushita
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan.,Department of Pharmacology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Syuuichirou Suzuki
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Shun Shimohama
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo, Hokkaido, Japan
| |
Collapse
|
24
|
George J, Wang J. Assay of Genome-Wide Transcriptome and Secreted Proteins on the Same Single Immune Cells by Microfluidics and RNA Sequencing. Anal Chem 2016; 88:10309-10315. [PMID: 27626628 DOI: 10.1021/acs.analchem.6b03214] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Given vast heterogeneity of immune cells, searching for gene expression and transcriptional networks belonging to specific cellular functions such as cytokine production has been challenging. To overcome this limitation, we developed a splittable single-cell microchip that integrates a high-density antibody array for cytokine protein detection, while the same single cells with protein profiles can be subsequently sequenced to obtain the genome-wide transcriptome. Combined with bioinformatics algorithms, we discovered a subgroup of highly coexpressed genes correlating with TNFα secretion in mouse macrophage cells. This technology and the data analysis may lead to an unprecedented understanding of regulation mechanisms of the immune system and have the potential to impact disease treatment and drug discovery.
Collapse
Affiliation(s)
- Justin George
- Multiplex Biotechnology Laboratory, Department of Chemistry, University at Albany, State University of New York , Albany, New York 12222, United States
| | - Jun Wang
- Multiplex Biotechnology Laboratory, Department of Chemistry, University at Albany, State University of New York , Albany, New York 12222, United States.,Cancer Research Center, University at Albany, State University of New York , Rensselaer, New York 12144, United States
| |
Collapse
|
25
|
Chronic IL-6 Administration Desensitizes IL-6 Response in Liver, Causes Hyperleptinemia and Aggravates Steatosis in Diet-Induced-Obese Mice. PLoS One 2016; 11:e0157956. [PMID: 27333268 PMCID: PMC4917096 DOI: 10.1371/journal.pone.0157956] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 06/07/2016] [Indexed: 02/06/2023] Open
Abstract
High-fat diet-induced obesity (DIO) is associated with fatty liver and elevated IL-6 circulating levels. IL-6 administration in rodents has yielded contradictory results regarding its effects on steatosis progression. In some models of fatty liver disease, high doses of human IL-6 ameliorate the liver steatosis, whereas restoration of IL-6 in DIO IL-6-/- mice up-regulates hepatic lipogenic enzymes and aggravates steatosis. We further examined the effects of chronic low doses of murine IL-6 on hepatic lipid metabolism in WT mice in DIO. IL-6 was delivered twice daily in C57BL/6J DIO mice for 15 days. The status and expression of IL-6-signalling mediators and targets were investigated in relation to the steatosis and lipid content in blood and in liver. IL-6 administration in DIO mice markedly raised circulating levels of lipids, glucose and leptin, elevated fat liver content and aggravated steatosis. Under IL-6 treatment there was hepatic Stat3 activation and increased gene expression of Socs3 and Tnf-alpha whereas the gene expression of endogenous IL-6, IL-6-receptor, Stat3, Cpt1 and the enzymes involved in lipogenesis was suppressed. These data further implicate IL-6 in fatty liver disease modulation in the context of DIO, and indicate that continuous stimulation with IL-6 attenuates the IL-6-receptor response, which is associated with high serum levels of leptin, glucose and lipids, the lowering levels of lipogenic and Cpt1 hepatic enzymes and with increased Tnf-alpha hepatic expression, a scenario evoking that observed in IL-6-/- mice exposed to DIO and in obese Zucker rats.
Collapse
|
26
|
Masamoto Y, Arai S, Sato T, Yoshimi A, Kubota N, Takamoto I, Iwakura Y, Yoshimura A, Kadowaki T, Kurokawa M. Adiponectin Enhances Antibacterial Activity of Hematopoietic Cells by Suppressing Bone Marrow Inflammation. Immunity 2016; 44:1422-33. [PMID: 27317261 DOI: 10.1016/j.immuni.2016.05.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 11/06/2015] [Accepted: 04/05/2016] [Indexed: 11/30/2022]
Abstract
Obesity has been shown to increase the morbidity of infections, however, the underlying mechanisms remain largely unknown. Here we demonstrate that obesity caused adiponectin deficiency in the bone marrow (BM), which led to an inflamed BM characterized by increased tumor necrosis factor (TNF) production from bone marrow macrophages. Hematopoietic stem and progenitor cells (HSPCs) chronically exposed to excessive TNF in obese marrow aberrantly expressed cytokine signaling suppressor SOCS3, impairing JAK-STAT mediated signal transduction and cytokine-driven cell proliferation. Accordingly, both obese and adiponectin-deficient mice showed attenuated clearance of infected Listeria monocytogenes, indicating that obesity or loss of adiponectin is critical for exacerbation of infection. Adiponectin treatment restored the defective HSPC proliferation and bacterial clearance of obese and adiponectin-deficient mice, affirming the importance of adiponectin against infection. Taken together, our findings demonstrate that obesity impairs hematopoietic response against infections through a TNF-SOCS3-STAT3 axis, highlighting adiponectin as a legitimate target against obesity-related infections.
Collapse
Affiliation(s)
- Yosuke Masamoto
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Transfusion Medicine, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Shunya Arai
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tomohiko Sato
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; Department of Transfusion Medicine, The University of Tokyo Hospital, Tokyo 113-8655, Japan
| | - Akihide Yoshimi
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Iseki Takamoto
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yoichiro Iwakura
- Division of Experimental Animal Immunology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan.
| |
Collapse
|
27
|
Dudek SE, Nitzsche K, Ludwig S, Ehrhardt C. Influenza A viruses suppress cyclooxygenase-2 expression by affecting its mRNA stability. Sci Rep 2016; 6:27275. [PMID: 27265729 PMCID: PMC4893666 DOI: 10.1038/srep27275] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 05/13/2016] [Indexed: 02/04/2023] Open
Abstract
Infection with influenza A viruses (IAV) provokes activation of cellular defence mechanisms contributing to the innate immune and inflammatory response. In this process the cyclooxygenase-2 (COX-2) plays an important role in the induction of prostaglandin-dependent inflammation. While it has been reported that COX-2 is induced upon IAV infection, in the present study we observed a down-regulation at later stages of infection suggesting a tight regulation of COX-2 by IAV. Our data indicate the pattern-recognition receptor RIG-I as mediator of the initial IAV-induced COX-2 synthesis. Nonetheless, during on-going IAV replication substantial suppression of COX-2 mRNA and protein synthesis could be detected, accompanied by a decrease in mRNA half-life. Interestingly, COX-2 mRNA stability was not only imbalanced by IAV replication but also by stimulation of cells with viral RNA. Our results reveal tristetraprolin (TTP), which is known to bind COX-2 mRNA and promote its rapid degradation, as regulator of COX-2 expression in IAV infection. During IAV replication and viral RNA accumulation TTP mRNA synthesis was induced, resulting in reduced COX-2 levels. Accordingly, the down-regulation of TTP resulted in increased COX-2 protein expression after IAV infection. These findings indicate a novel IAV-regulated cellular mechanism, contributing to the repression of host defence and therefore facilitating viral replication.
Collapse
Affiliation(s)
- Sabine Eva Dudek
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
| | - Katja Nitzsche
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
| | - Stephan Ludwig
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
- Cluster of Excellence Cells in Motion, University of Muenster, Muenster, Germany
| | - Christina Ehrhardt
- Institute of Molecular Virology (IMV), Centre for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University Muenster, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
- Cluster of Excellence Cells in Motion, University of Muenster, Muenster, Germany
| |
Collapse
|
28
|
Shen M, Shi H. Estradiol and Estrogen Receptor Agonists Oppose Oncogenic Actions of Leptin in HepG2 Cells. PLoS One 2016; 11:e0151455. [PMID: 26982332 PMCID: PMC4794158 DOI: 10.1371/journal.pone.0151455] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 02/28/2016] [Indexed: 11/19/2022] Open
Abstract
Obesity is a significant risk factor for certain cancers, including hepatocellular carcinoma (HCC). Leptin, a hormone secreted by white adipose tissue, precipitates HCC development. Epidemiology data show that men have a much higher incidence of HCC than women, suggesting that estrogens and its receptors may inhibit HCC development and progression. Whether estrogens antagonize oncogenic action of leptin is uncertain. To investigate potential inhibitory effects of estrogens on leptin-induced HCC development, HCC cell line HepG2 cells were treated with leptin in combination with 17 β-estradiol (E2), estrogen receptor-α (ER-α) selective agonist PPT, ER-β selective agonist DPN, or G protein-coupled ER (GPER) selective agonist G-1. Cell number, proliferation, and apoptosis were determined, and leptin- and estrogen-related intracellular signaling pathways were analyzed. HepG2 cells expressed a low level of ER-β mRNA, and leptin treatment increased ER-β expression. E2 suppressed leptin-induced HepG2 cell proliferation and promoted cell apoptosis in a dose-dependent manner. Additionally E2 reversed leptin-induced STAT3 and leptin-suppressed SOCS3, which was mainly achieved by activation of ER-β. E2 also enhanced ERK via activating ER-α and GPER and activated p38/MAPK via activating ER-β. To conclude, E2 and its receptors antagonize the oncogenic actions of leptin in HepG2 cells by inhibiting cell proliferation and stimulating cell apoptosis, which was associated with reversing leptin-induced changes in SOCS3/STAT3 and increasing p38/MAPK by activating ER-β, and increasing ERK by activating ER-α and GPER. Identifying roles of different estrogen receptors would provide comprehensive understanding of estrogenic mechanisms in HCC development and shed light on potential treatment for HCC patients.
Collapse
Affiliation(s)
- Minqian Shen
- Department of Biology, Cell, Molecular, and Structural Biology, Miami University, Oxford, Ohio, United States of America
| | - Haifei Shi
- Department of Biology, Cell, Molecular, and Structural Biology, Miami University, Oxford, Ohio, United States of America
- * E-mail:
| |
Collapse
|
29
|
Nita-Lazar M, Banerjee A, Feng C, Vasta GR. Galectins regulate the inflammatory response in airway epithelial cells exposed to microbial neuraminidase by modulating the expression of SOCS1 and RIG1. Mol Immunol 2015; 68:194-202. [PMID: 26355912 PMCID: PMC4624043 DOI: 10.1016/j.molimm.2015.08.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 01/12/2023]
Abstract
Influenza patients frequently display increased susceptibility to Streptococcus pneumoniae co-infection and sepsis, the prevalent cause of mortality during influenza pandemics. However, the detailed mechanisms by which an influenza infection predisposes patients to suffer pneumococcal pneumonia are not fully understood. A murine model for influenza infection closely reflects the observations in human patients, since if the animals that have recovered from influenza A virus (IAV) sublethal infection are challenged with S. pneumoniae, they undergo a usually fatal uncontrolled cytokine response. We have previously demonstrated both in vitro and in vivo that the expression and secretion of galectin-1 (Gal1) and galectin-3 (Gal3) are modulated during IAV infection, and that the viral neuraminidase unmasks galactosyl moieties in the airway epithelia. In this study we demonstrate in vitro that the binding of secreted Gal1 and Gal3 to the epithelial cell surface modulates the expression of SOCS1 and RIG1, and activation of ERK, AKT or JAK/STAT1 signaling pathways, leading to a disregulated expression and release of pro-inflammatory cytokines. Our results suggest that the activity of the viral and pneumococcal neuraminidases on the surface of the airway epithelial cells function as a "danger signal" that leads to rapid upregulation of SOCS1 expression to prevent an uncontrolled inflammatory response. The binding of extracellular Gal1 or Gal3 to the galactosyl moieties unmasked on the surface of airway epithelial cells can either "fine-tune" or severely disregulate this process, respectively, the latter potentially leading to hypercytokinemia.
Collapse
Affiliation(s)
- Mihai Nita-Lazar
- Department of Microbiology and Immunology, University of Maryland School of Medicine, and Institute of Marine and Environmental Technology, Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA
| | - Aditi Banerjee
- Department of Microbiology and Immunology, University of Maryland School of Medicine, and Institute of Marine and Environmental Technology, Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA
| | - Chiguang Feng
- Department of Microbiology and Immunology, University of Maryland School of Medicine, and Institute of Marine and Environmental Technology, Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA
| | - Gerardo R Vasta
- Department of Microbiology and Immunology, University of Maryland School of Medicine, and Institute of Marine and Environmental Technology, Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, USA.
| |
Collapse
|
30
|
López-Yoldi M, Moreno-Aliaga MJ, Bustos M. Cardiotrophin-1: A multifaceted cytokine. Cytokine Growth Factor Rev 2015; 26:523-32. [PMID: 26188636 DOI: 10.1016/j.cytogfr.2015.07.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 07/01/2015] [Indexed: 10/23/2022]
Abstract
Cardiotrophin-1 (CT-1) is a member of the gp130 family of cytokines that have pleiotropic functions on different tissues and cell types. Although many effects of CT-1 have been described on the heart, there is an extensive research showing important protective effects in other organs such as liver, kidney or nervous system. Recently, several studies have pointed out that CT-1 might also play a key role in the regulation of body weight and intermediate metabolism. This paper will review many aspects of CT-1 physiological role in several organs and discuss data for consideration in therapeutic approaches.
Collapse
Affiliation(s)
- Miguel López-Yoldi
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, University of Navarra, Pamplona, Spain
| | - María J Moreno-Aliaga
- Department of Nutrition, Food Science and Physiology, University of Navarra, Pamplona, Spain; Centre for Nutrition Research, University of Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Matilde Bustos
- Area of Hepatology and Gene Therapy, CIMA (Center for Applied Medical Research) University of Navarra, Pamplona, Spain.
| |
Collapse
|
31
|
Green AC, Poulton IJ, Vrahnas C, Häusler KD, Walkley CR, Wu JY, Martin TJ, Gillespie MT, Chandraratna RAS, Quinn JMW, Sims NA, Purton LE. RARγ is a negative regulator of osteoclastogenesis. J Steroid Biochem Mol Biol 2015; 150:46-53. [PMID: 25800721 DOI: 10.1016/j.jsbmb.2015.03.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/03/2015] [Accepted: 03/17/2015] [Indexed: 01/14/2023]
Abstract
Vitamin A is known to influence post-natal bone content, with excess intake being associated with reduced bone mineral density and increased fracture risk. Despite this, the roles retinoids play in regulating osteoclastogenesis, particularly in vivo, remain unresolved. This study therefore aimed to determine the effect of loss of retinoic acid receptors (RAR)α or RARγ on bone mass (analyzed by histomorphometry and dual-energy X-ray absorptiometry) and osteoclastogenesis in mice in vivo. RARγ null mice had significantly less trabecular bone at 8 weeks of age compared to wildtype littermates. In contrast, no change in trabecular bone mass was detected in RARα null mice at this age. Further histomorphometric analysis revealed a significantly greater osteoclast surface in bones from 8-week-old RARγ null male mice. This in vivo effect was cell lineage autonomous, and was associated with increased osteoclastogenesis in vitro from hematopoietic cells obtained from 8-week-old RARγ null male mice. The use of highly selective agonists in RANKL-induced osteoclast differentiation of wild type mouse whole bone marrow cells and RAW264.7 cells in vitro showed a stronger inhibitory effect of RARγ than RARα agonists, suggesting that RARγ is a more potent inhibitor of osteoclastogenesis. Furthermore, NFAT activation was also more strongly inhibited by RARγ than RARα agonists. While RARα and RARγ antagonists did not significantly affect osteoclast numbers in vitro, larger osteoclasts were observed in cultures stimulated with the antagonists, suggesting increased osteoclast fusion. Further investigation into the effect of retinoids in vivo revealed that oral administration of 5mg/kg/day ATRA for 10 days protected against bone loss induced by granulocyte colony-stimulating factor (G-CSF) by inhibiting the pro-osteoclastogenic action of G-CSF. Collectively, our data indicates a physiological role for RARγ as a negative regulator of osteoclastogenesis in vivo and in vitro, and reveals distinct influences of RARα and RARγ in bone structure regulation.
Collapse
Affiliation(s)
- Alanna C Green
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3065, Australia
| | | | - Christina Vrahnas
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3065, Australia
| | - Karl D Häusler
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia
| | - Carl R Walkley
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3065, Australia
| | - Joy Y Wu
- Division of Endocrinology, Stanford University School of Medicine, CA 94305, USA
| | - T John Martin
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3065, Australia
| | - Matthew T Gillespie
- MIMR-PHI Institute, Monash Medical Centre, Clayton, Victoria 3168, Australia
| | | | - Julian M W Quinn
- MIMR-PHI Institute, Monash Medical Centre, Clayton, Victoria 3168, Australia; The Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010 Australia
| | - Natalie A Sims
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3065, Australia
| | - Louise E Purton
- St. Vincent's Institute, Fitzroy, Victoria 3065, Australia; Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Victoria 3065, Australia.
| |
Collapse
|
32
|
Fu X, Ren L, Chen J, Liao K, Fu Y, Qian X, Xiao J. Characterization of the roles of suppressor of cytokine signaling-3 in prostate cancer development and progression. Asia Pac J Clin Oncol 2015; 11:106-13. [PMID: 25899712 DOI: 10.1111/ajco.12357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2015] [Indexed: 01/01/2023]
Abstract
As negative feedback regulators of cytokine signaling, suppressor of cytokine signaling proteins are induced by interleukins and various peptide hormones and may prevent sustained activation of signaling pathways. In particular, suppressor of cytokine signaling-3 (SOCS-3) plays pivotal roles in the development and progression of various cancers and exerts pleiotropic effects on cell proliferation and apoptosis. In recent years, abnormal expression of SOCS-3 and its multiple functions have been extensively investigated in human carcinomas, particularly in prostate cancer. SOCS-3 can act as an oncogene or a tumor suppressor depending on the cellular context. In this review, we focus on the role of SOCS-3 in prostate cancer development and prognosis, as well as the potential of SOCS-3 as a therapeutic target and diagnostic marker.
Collapse
Affiliation(s)
- Xian Fu
- Department of Urology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
33
|
Mauer J, Denson JL, Brüning JC. Versatile functions for IL-6 in metabolism and cancer. Trends Immunol 2015; 36:92-101. [PMID: 25616716 DOI: 10.1016/j.it.2014.12.008] [Citation(s) in RCA: 276] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 12/26/2014] [Accepted: 12/26/2014] [Indexed: 12/16/2022]
Abstract
Owing to its abundance in inflammatory settings, interleukin IL-6 is frequently viewed as a proinflammatory cytokine, with functions that parallel those of tumor necrosis factor (TNF) and IL-1β in the context of inflammation. However, accumulating evidence points to a broader role for IL-6 in a variety of (patho)physiological conditions, including functions related to the resolution of inflammation. We review recent findings on the complex biological functions governed by IL-6 signaling, focusing on its role in inflammation-associated cancer and metabolic disorders such as obesity and type 2 diabetes mellitus (T2DM). We propose that the anti-inflammatory functions of IL-6 may extend to multiple settings and cell types, and suggest that these dimensions should be incorporated in therapeutic approaches to these diseases. Finally, we outline important areas of inquiry towards understanding this pleiotropic cytokine.
Collapse
Affiliation(s)
- Jan Mauer
- Max Planck Institute for Metabolism Research, Cologne, Germany.
| | - Jesse L Denson
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Jens C Brüning
- Max Planck Institute for Metabolism Research, Cologne, Germany.
| |
Collapse
|
34
|
Ehlting C, Böhmer O, Hahnel MJ, Thomas M, Zanger UM, Gaestel M, Knoefel WT, Schulte Am Esch J, Häussinger D, Bode JG. Oncostatin M regulates SOCS3 mRNA stability via the MEK-ERK1/2-pathway independent of p38(MAPK)/MK2. Cell Signal 2015; 27:555-67. [PMID: 25562430 DOI: 10.1016/j.cellsig.2014.12.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 12/13/2014] [Accepted: 12/28/2014] [Indexed: 12/22/2022]
Abstract
The induction of suppressor of cytokine signalling (SOCS)3 expression context dependently involves regulation of SOCS3 transcript stability as previously demonstrated for MAPK activated protein kinase (MK)2-dependent regulation of SOCS3 expression by TNFα (Ehlting et al., 2007). In how far the IL-6-type cytokine OSM, which in contrast to IL-6 is a strong activator of p38(MAPK)/MK2 signalling, also involves regulation of transcript stability and activation of MK2 to induce SOCS3 expression is unclear. In contrast to IL-6, OSM induces SOCS3 expression in murine fibroblasts and in primary human and murine hepatocytes, but not in macrophages because the latter lack the OSM receptor (OSMR)β subunit. Evidence is provided that regulation of OSM-induced expression of SOCS3 involves MEK1- and Erk1/2-mediated stabilization of the SOCS3 transcript. Consistently, OSM-induced stabilization of the SOCS3 transcript is impaired in the presence of inhibitors that specifically block activation of MEK1/2 (U0126) and ERK1/2 (FR180204) or upon knock-down of ERK1/2 expression using specific siRNA. As a potential target site that integrates the stability regulating effect of OSM and OSM-induced activation of MEK1/2 and ERK1/2 a region containing three copies of a pentameric AUUUA motif located within position 2422 and 2541 in closed proximity to the 3' UTR of the SOCS3 transcript has been identified. Unexpectedly, activation of the p38(MAPK)/MK2 pathway, which apart from STAT3 and ERK1/2, is also strongly activated by OSM in human and murine hepatocytes and murine fibroblasts is dispensable for stabilization of the SOCS3 transcript as suggested from inhibitor studies using the p38(MAPK) inhibitor SB203580 or from the analysis of MK2-deficient hepatocytes. However, analysis of MK2-deficient macrophages and hepatocytes revealed that, although MK2 is dispensable for regulation of OSM-induced SOCS3 expression, MK2 is essential for LPS-induced OSM production in macrophages and limits the overall availability of the OSMRβ subunit in hepatocytes. Thus MK2 plays a role for the induction and sensing of OSM-mediated intercellular signalling between macrophages and hepatocytes during LPS-induced inflammation.
Collapse
Affiliation(s)
- Christian Ehlting
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Oliver Böhmer
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Maximilian J Hahnel
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Maria Thomas
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
| | - Ulrich M Zanger
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, 70376 Stuttgart, Germany
| | - Matthias Gaestel
- Institute of Physiological Chemistry, Hannover Medical School, 30623 Hannover, Germany
| | - Wolfram T Knoefel
- Department of Surgery (A), Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Jan Schulte Am Esch
- Department of Surgery (A), Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Dieter Häussinger
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Johannes G Bode
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, University Hospital, Heinrich Heine University of Düsseldorf, Moorenstrasse 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
35
|
Cates AM, Holden VI, Myers EM, Smith CK, Kaplan MJ, Kahlenberg JM. Interleukin 10 hampers endothelial cell differentiation and enhances the effects of interferon α on lupus endothelial cell progenitors. Rheumatology (Oxford) 2014; 54:1114-23. [PMID: 25416712 DOI: 10.1093/rheumatology/keu431] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE SLE is an autoimmune disease characterized by autoantibody generation, organ damage and an increased risk of cardiovascular disease. Generally considered an anti-inflammatory cytokine, IL-10 is increased in SLE and correlates with poor cardiovascular outcomes in the general population. The aim of this study was to explore the putative role of IL-10 in modulating endothelial function in SLE by examining the effects of this cytokine on endothelial progenitor cell/circulating angiogenic cell (EPC/CAC) differentiation. METHODS Human and murine control and lupus EPCs/CACs were differentiated into mature endothelial cells (ECs) in the presence or absence of graded concentrations of recombinant IL-10 with or without recombinant IFN-α or a neutralizing antibody to IL-10. IL-10-deficient mice were examined to assess the role of this cytokine in type I IFN-mediated inhibition of EC differentiation and neo-angiogenesis using an in vivo Matrigel plug assay. Serum IL-10 concentrations were measured via ELISA. RESULTS IL-10 hampers EC differentiation in a dose-dependent manner. In murine EPC cultures, IL-10 is required to observe the inhibitory effects of type I IFNs on EPC function and neo-angiogenesis. In human SLE EPC/CAC cultures, neutralization of IL-10 significantly improved the differentiation of EPCs, and IL-10 enhanced type I IFN-mediated EPC/CAC dysfunction. The presence of IL-10 in serum inversely correlated with EPC/CAC function in SLE but not in control cells. CONCLUSION IL-10 interferes with endothelial differentiation and may enhance the effects of type I IFN on vascular repair in SLE. IL-10 may be a relevant target for improving cardiovascular risk in SLE.
Collapse
Affiliation(s)
- Alyssa M Cates
- Department of Internal Medicine, Division of Rheumatology, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI and Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Victoria I Holden
- Department of Internal Medicine, Division of Rheumatology, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI and Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Emily M Myers
- Department of Internal Medicine, Division of Rheumatology, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI and Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Carolyne K Smith
- Department of Internal Medicine, Division of Rheumatology, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI and Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Disorders, National Institutes of Health, Bethesda, MD, USA
| | - Mariana J Kaplan
- Department of Internal Medicine, Division of Rheumatology, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI and Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Disorders, National Institutes of Health, Bethesda, MD, USA
| | - J Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI and Systemic Autoimmunity Branch, Intramural Research Program, National Institute of Arthritis and Musculoskeletal and Skin Disorders, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
36
|
Pimentel GD, Ganeshan K, Carvalheira JBC. Hypothalamic inflammation and the central nervous system control of energy homeostasis. Mol Cell Endocrinol 2014; 397:15-22. [PMID: 24952114 DOI: 10.1016/j.mce.2014.06.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 06/07/2014] [Accepted: 06/08/2014] [Indexed: 02/07/2023]
Abstract
The control of energy homeostasis relies on robust neuronal circuits that regulate food intake and energy expenditure. Although the physiology of these circuits is well understood, the molecular and cellular response of this program to chronic diseases is still largely unclear. Hypothalamic inflammation has emerged as a major driver of energy homeostasis dysfunction in both obesity and anorexia. Importantly, this inflammation disrupts the action of metabolic signals promoting anabolism or supporting catabolism. In this review, we address the evidence that favors hypothalamic inflammation as a factor that resets energy homeostasis in pathological states.
Collapse
Affiliation(s)
- Gustavo D Pimentel
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Kirthana Ganeshan
- Cardiovascular Research Institute, University of California, San Francisco, CA 94158-9001, United States
| | - José B C Carvalheira
- Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
37
|
Meng A, Zhang X, Shi Y. Role of p38 MAPK and STAT3 in lipopolysaccharide-stimulated mouse alveolar macrophages. Exp Ther Med 2014; 8:1772-1776. [PMID: 25371731 PMCID: PMC4218692 DOI: 10.3892/etm.2014.2023] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 10/02/2014] [Indexed: 12/12/2022] Open
Abstract
Excessive production of inflammatory mediators is an important feature of inflammatory lung disease. In macrophages, mitogen-activated protein kinase (MAPK) and signal transducer and activator of transcription-3 (STAT3) are crucial mediators for the production of proinflammatory cytokines. In the present study, the role of MAPK and STAT3 on tumor necrosis factor (TNF)-α and interleukin (IL)-10 production was investigated in mouse alveolar macrophages. The levels of TNF-α and IL-10 in lipopolysaccharide (LPS; 100 ng/ml)-stimulated MH-S cell lines were measured by an enzyme-linked immunosorbent assay, with or without p38 inhibitor (SB203580; 5, 10 or 15 μM) intervention. Phosphorylated STAT3 (p-STAT3) expression was examined by western blot analysis and immunocytochemistry following LPS stimulation for 15 or 30 min. Antibodies against STAT3 were used to verify comparable sample loading. Cells stimulated with LPS showed significantly increased levels of p-STAT3 protein (P<0.05) when compared with the baseline levels. TNF-α and IL-10 protein levels also increased following LPS stimulation (P<0.05). By contrast, treatment with the p38 inhibitor, SB203580, decreased the levels of p-STAT3, TNF-α and IL-10 (P<0.05) following LPS stimulation. SB203580 was shown to inhibit LPS-stimulated TNF-α expression (P<0.05) in a concentration-dependent manner, reaching significance at a concentration of 10 μM. However, the inhibition of IL-10 expression was not concentration-dependent. Therefore, LPS-stimulated overproduction of TNF-α and IL-10 is mediated at least partially by the MAPK pathway. Inhibition of p38 prevented LPS-induced STAT3 phosphorylation, indicating an interaction between the STAT3 and MAPK signaling pathways.
Collapse
Affiliation(s)
- Aihong Meng
- Respiratory Division, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiaopeng Zhang
- Graduate School, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China ; Department of Thoracic Surgery, Hebei Province General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Yuna Shi
- Respiratory Division, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
38
|
Walker DG, Whetzel AM, Lue LF. Expression of suppressor of cytokine signaling genes in human elderly and Alzheimer's disease brains and human microglia. Neuroscience 2014; 302:121-37. [PMID: 25286386 DOI: 10.1016/j.neuroscience.2014.09.052] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 09/23/2014] [Accepted: 09/23/2014] [Indexed: 12/16/2022]
Abstract
Multiple cellular systems exist to prevent uncontrolled inflammation in brain tissues; the suppressor of cytokine signaling (SOCS) proteins have key roles in these processes. SOCS proteins are involved in restricting cellular signaling pathways by enhancing the degradation of activated receptors and removing the stimuli for continued activation. There are eight separate SOCS genes that code for proteins with similar structures and properties. All SOCS proteins can reduce signaling of activated transcription factors Janus kinase (JAK) and signal transducer and activator of transcription (STAT), but they also regulate many other signaling pathways. SOCS-1 and SOCS-3 have particular roles in regulating inflammatory processes. Chronic inflammation is a key feature of the pathology present in Alzheimer's disease (AD)-affected brains resulting from responses to amyloid plaques or neurofibrillary tangles, the pathological hallmarks of AD. The goal of this study was to examine SOCS gene expression in human non-demented (ND) and AD brains and in human brain-derived microglia to determine if AD-related pathology resulted in a deficit of these critical molecules. We demonstrated that SOCS-1, SOCS-2, SOCS-3 and cytokine-inducible SH2 containing protein (CIS) mRNA expression was increased in amyloid beta peptide (Aβ)- and inflammatory-stimulated microglia, while SOCS-6 mRNA expression was decreased by both types of treatments. Using human brain samples from the temporal cortex from ND and AD cases, SOCS-1 through SOCS-7 and CIS mRNA and SOCS-1 through SOCS-7 protein could be detected constitutively in ND and AD human brain samples. Although, the expression of key SOCS genes did not change to a large extent as a result of AD pathology, there were significantly increased levels of SOCS-2, SOCS-3 and CIS mRNA and increased protein levels of SOCS-4 and SOCS-7 in AD brains. In summary, there was no evidence of a deficit of these key inflammatory regulating proteins in aged or AD brains.
Collapse
Affiliation(s)
- D G Walker
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| | - A M Whetzel
- Laboratory of Neuroinflammation, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| | - L-F Lue
- Laboratory of NeuroRegeneration, Banner Sun Health Research Institute, Sun City, AZ 85351, USA.
| |
Collapse
|
39
|
Martel CA, Mamedova LK, Minton JE, Jones ML, Carroll JA, Bradford BJ. Continuous low-dose infusion of tumor necrosis factor alpha in adipose tissue elevates adipose tissue interleukin 10 abundance and fails to alter metabolism in lactating dairy cows. J Dairy Sci 2014; 97:4897-906. [DOI: 10.3168/jds.2013-7777] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/17/2014] [Indexed: 12/14/2022]
|
40
|
Honke N, Ohl K, Wiener A, Bierwagen J, Peitz J, Di Fiore S, Fischer R, Wagner N, Wüller S, Tenbrock K. The p38-Mediated Rapid Down-Regulation of Cell Surface gp130 Expression Impairs Interleukin-6 Signaling in the Synovial Fluid of Juvenile Idiopathic Arthritis Patients. Arthritis Rheumatol 2014; 66:470-8. [DOI: 10.1002/art.38245] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 10/17/2013] [Indexed: 11/10/2022]
Affiliation(s)
| | - Kim Ohl
- RWTH Aachen University; Aachen Germany
| | | | | | | | - Stefano Di Fiore
- Fraunhofer Institute for Molecular Biology and Applied Ecology; Aachen Germany
| | - Rainer Fischer
- Fraunhofer Institute for Molecular Biology and Applied Ecology; Aachen Germany
| | | | | | | |
Collapse
|
41
|
Hsu CK, Lee IT, Lin CC, Hsiao LD, Yang CM. Nox2/ROS-dependent human antigen R translocation contributes to TNF-α-induced SOCS-3 expression in human tracheal smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2014; 306:L521-33. [PMID: 24414258 DOI: 10.1152/ajplung.00274.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Elevated levels of TNF-α have been detected in the airway fluids, which may induce upregulation of inflammatory proteins. Suppressors of cytokine signaling (SOCS)-3 proteins can be induced by various cytokines and negatively regulated inflammatory responses. Although TNF-α has been shown to induce SOCS-3 expression, the mechanisms underlying TNF-α-induced SOCS-3 expression in human tracheal smooth muscle cells (HTSMCs) remain unclear. Here, we showed that TNF-α induced SOCS-3 expression, which was inhibited by pretreatment with the inhibitor of transcription level (actinomycin D), translation level (cycloheximide), JNK1/2 (SP600125), MEK1/2 (U0126), NADPH oxidase (Nox; apocynin and diphenyleneiodonium chloride), or reactive oxygen species (ROS; N-acetyl-l-cysteine) and transfection with siRNA of JNK1, p47(phox), p42, Nox2, or human antigen R (HuR). In addition, TNF-α-stimulated JNK1/2 and p42/p44 MAPK phosphorylation, Nox activation, and ROS generation were inhibited by pretreatment with U0126 or SP600125 and transfection with siRNA of JNK1 or p42. We further showed that TNF-α markedly induced HuR protein expression and translocation from the nucleus to the cytosol, which could stabilize SOCS-3 mRNA. Moreover, TNF-α-enhanced HuR translocation was reduced by transfection with siRNA of p42, JNK1, or p47(phox). These results suggested that TNF-α induces SOCS-3 protein expression and mRNA stabilization via a TNFR1/JNK1/2, p42/p44 MAPK/Nox2/ROS-dependent HuR signaling in HTSMCs. Lipopolysaccharide (LPS) has been shown to play a key role in inflammation via induction of adhesion molecules and then causes airway and lung injury. Moreover, we also demonstrated that overexpression of SOCS-3 protects against LPS-induced adhesion molecules expression and airway inflammation.
Collapse
Affiliation(s)
- Chih-Kai Hsu
- Dept. of Pharmacology, Chang Gung Univ., 259 Wen-Hwa 1st Rd., Kwei-San, Tao-Yuan, Taiwan.
| | | | | | | | | |
Collapse
|
42
|
Wang T, Secombes CJ. The cytokine networks of adaptive immunity in fish. FISH & SHELLFISH IMMUNOLOGY 2013; 35:1703-1718. [PMID: 24036335 DOI: 10.1016/j.fsi.2013.08.030] [Citation(s) in RCA: 213] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 08/30/2013] [Accepted: 08/31/2013] [Indexed: 05/28/2023]
Abstract
Cytokines, produced at the site of entry of a pathogen, drive inflammatory signals that regulate the capacity of resident and newly arrived phagocytes to destroy the invading pathogen. They also regulate antigen presenting cells (APCs), and their migration to lymph nodes to initiate the adaptive immune response. When naive CD4+ T cells recognize a foreign antigen-derived peptide presented in the context of major histocompatibility complex class II on APCs, they undergo massive proliferation and differentiation into at least four different T-helper (Th) cell subsets (Th1, Th2, Th17, and induced T-regulatory (iTreg) cells in mammals. Each cell subset expresses a unique set of signature cytokines. The profile and magnitude of cytokines produced in response to invasion of a foreign organism or to other danger signals by activated CD4+ T cells themselves, and/or other cell types during the course of differentiation, define to a large extent whether subsequent immune responses will have beneficial or detrimental effects to the host. The major players of the cytokine network of adaptive immunity in fish are described in this review with a focus on the salmonid cytokine network. We highlight the molecular, and increasing cellular, evidence for the existence of T-helper cells in fish. Whether these cells will match exactly to the mammalian paradigm remains to be seen, but the early evidence suggests that there will be many similarities to known subsets. Alternative or additional Th populations may also exist in fish, perhaps influenced by the types of pathogen encountered by a particular species and/or fish group. These Th cells are crucial for eliciting disease resistance post-vaccination, and hopefully will help resolve some of the difficulties in producing efficacious vaccines to certain fish diseases.
Collapse
Affiliation(s)
- Tiehui Wang
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, UK.
| | | |
Collapse
|
43
|
Hong S, Li R, Xu Q, Secombes CJ, Wang T. Two types of TNF-α exist in teleost fish: phylogeny, expression, and bioactivity analysis of type-II TNF-α3 in rainbow trout Oncorhynchus mykiss. THE JOURNAL OF IMMUNOLOGY 2013; 191:5959-72. [PMID: 24244011 DOI: 10.4049/jimmunol.1301584] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
TNF-α is a cytokine involved in systemic inflammation and regulation of immune cells. It is produced chiefly by activated macrophages as a membrane or secreted form. In rainbow trout, two TNF-α molecules were described previously. In this article, we report a third TNF-α (TNF-α3) that has only low identities to known trout molecules. Phylogenetic tree and synteny analyses of trout and other fish species suggest that two types (named I and II) of TNF-α exist in teleost fish. The fish type-II TNF-α has a short stalk that may impact on its enzymatic release or restrict it to a membrane-bound form. The constitutive expression of trout TNF-α3 was generally lower than the other two genes in tissues and cell lines, with the exception of the macrophage RTS-11 cell line, in which expression was higher. Expression of all three TNF-α isoforms could be modulated by crude LPS, peptidoglycan, polyinosinic:polycytidylic acid, and rIFN-γ in cell lines and primary macrophages, as well as by bacterial and viral infections. TNF-α3 is the most responsive gene at early time points post-LPS stimulation and can be highly induced by the T cell-stimulant PHA, suggesting it is a particularly important TNF-α isoform. rTNF-α3 produced in CHO cells was bioactive in different cell lines and primary macrophages. In the latter, it induced the expression of proinflammatory cytokines (IL-1β, IL-6, IL-8, IL-17C, and TNF-αs), negative regulators (SOCS1-3, TGF-β1b), antimicrobial peptides (cathelicidin-1 and hepcidin), and the macrophage growth factor IL-34, verifying its key role in the inflammatory cytokine network and macrophage biology of fish.
Collapse
Affiliation(s)
- Suhee Hong
- Scottish Fish Immunology Research Centre, School of Biological Sciences, University of Aberdeen, Aberdeen AB24 2TZ, United Kingdom
| | | | | | | | | |
Collapse
|
44
|
Chhabra JK, Chattopadhyay B, Paul BN. SOCS3 dictates the transition of divergent time-phased events in granulocyte TNF-α signaling. Cell Mol Immunol 2013; 11:105-6. [PMID: 24037182 DOI: 10.1038/cmi.2013.36] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 07/02/2013] [Accepted: 07/04/2013] [Indexed: 11/09/2022] Open
Abstract
Tumor-necrosis factor-α (TNF-α)-driven nuclear factor-κB (NF-κB) activation and apoptosis are opposing pathways; the growing recognition of these conflicting roles of TNF-α is perplexing. Here, we show that inflammation and apoptosis are time-phased events following TNF-α signaling and that emergence of suppressor of cytokine signaling 3 (SOCS3) expression limits the ongoing NF-κB activation and promotes apoptosis; further, we suggest an altered view of how inflammatory diseases are initiated and sustained. In vitro, TNF-α (50 ng/ml) induced granulocyte SOCS3 protein, inhibited nuclear accumulation of the p65NF-κB subunit and enhanced apoptosis, as shown by DNA laddering, annexin V positivity, and overexpression of caspase-3 and Bax in the late phase, whereas the early phase was marked by NF-κB activation. Conversely, SOCS3 knockdown by small interfering RNA (siRNA) inhibited granulocyte apoptosis and enhanced nuclear accumulation of p65 and 5' lipooxygenase expression in the late phase of TNF-α signaling. As apoptosis is associated with SOCS3 abundance, we suggest that these divergent TNF-α-driven events are time-phased, interconnected, opposing control mechanisms and one of the central features through which the immune system resolves pulmonary inflammation. Dysregulation may initiate mucosal inflammation, thus changing the landscape of asthma therapy.
Collapse
Affiliation(s)
- Jasmeet Kaur Chhabra
- Immunobiology Division, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | | | - Bhola Nath Paul
- Immunobiology Division, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| |
Collapse
|
45
|
SOCS3 expression correlates with severity of inflammation, expression of proinflammatory cytokines, and activation of STAT3 and p38 MAPK in LPS-induced inflammation in vivo. Mediators Inflamm 2013; 2013:650812. [PMID: 24078776 PMCID: PMC3775441 DOI: 10.1155/2013/650812] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/27/2013] [Accepted: 07/11/2013] [Indexed: 01/09/2023] Open
Abstract
SOCS3 is an inducible endogenous negative regulator of JAK/STAT pathway, which is relevant in inflammatory conditions. We used a model of LPS-induced periodontal disease in rats to correlate SOCS3 expression with the inflammatory status. In vitro we used a murine macrophage cell line to assess the physical interaction between SOCS3 and STAT3 by coimmunoprecipitation. 30 ug of LPS from Escherichia coli were injected in the gingival tissues on the palatal aspect of first molars of the animals 3x/week for up to 4 weeks. Control animals were injected with the vehicle (PBS). The rats were sacrificed at 7, 15, and 30 days. Inflammation and gene expression were assessed by stereometric analysis, immunohistochemistry, RT-qPCR, and western blot. LPS injections increased inflammation, paralleled by an upregulation of SOCS3, of the proinflammatory cytokines IL-1 β , IL-6, and TNF- α and increased phosphorylation of STAT3 and p38 MAPK. SOCS3 expression accompanied the severity of inflammation and the expression of proinflammatory cytokines, as well as the activation status of STAT3 and p38 MAPK. LPS stimulation in a macrophage cell line in vitro induced transient STAT3 activation, which was inversely correlated with a dynamic physical interaction with SOCS3, suggesting that this may be a mechanism for SOCS3 regulatory function.
Collapse
|
46
|
White CA, Nicola NA. SOCS3: An essential physiological inhibitor of signaling by interleukin-6 and G-CSF family cytokines. JAKSTAT 2013; 2:e25045. [PMID: 24416642 PMCID: PMC3876435 DOI: 10.4161/jkst.25045] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022] Open
Abstract
SOCS3 is an inducible negative feedback inhibitor of cytokine signaling. Conditional deletion of SOCS3 in mice using the Cre-lox system has now been applied to a range of cell types in the steady-state and under inflammatory, pathogenic, or tumorigenic stress, with the resulting phenotypes demonstrating the effects of SOCS3 in physiological and disease contexts. Together with recent structural and biochemical studies on the mechanisms of SOCS3 binding to cytokine receptors and associated kinases, we now have a better understanding of the non-redundant roles of SOCS3 in the inhibition of cytokine signaling via the receptors gp130, G-CSFR, leptinR, and IL-12Rβ. This review discusses the known functional activities of SOCS3 in fertility and development, inflammation, innate and adaptive immunity, and malignancy as determined by genetic studies in mice.
Collapse
Affiliation(s)
- Christine A White
- Walter and Eliza Hall Institute of Medical Research; Parkville, VIC Australia ; Department of Medical Biology; University of Melbourne; Parkville, VIC Australia
| | - Nicos A Nicola
- Walter and Eliza Hall Institute of Medical Research; Parkville, VIC Australia ; Department of Medical Biology; University of Melbourne; Parkville, VIC Australia
| |
Collapse
|
47
|
van den Brand BT, Abdollahi-Roodsaz S, Vermeij EA, Bennink MB, Arntz OJ, Rothlin CV, van den Berg WB, van de Loo FAJ. Therapeutic efficacy of Tyro3, Axl, and Mer tyrosine kinase agonists in collagen-induced arthritis. ACTA ACUST UNITED AC 2013. [PMID: 23203851 DOI: 10.1002/art.37786] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Hyperactivation of innate immunity by Toll-like receptors (TLRs) can contribute to the development of autoinflammatory or autoimmune diseases. This study evaluated the activation of Tyro3, Axl, Mer (TAM) receptors, physiologic negative regulators of TLRs, by their agonists, growth arrest-specific protein 6 (GAS-6) and protein S, in the prevention of collagen-induced arthritis (CIA). METHODS Adenoviruses overexpressing GAS-6 and protein S were injected intravenously or intraarticularly into mice during CIA. Splenic T helper cell subsets from intravenously injected mice were studied by flow cytometry, and the knee joints of mice injected intravenously and intraarticularly were assessed histologically. Synovium from mice injected intraarticularly was evaluated for cytokine and suppressor of cytokine signaling (SOCS) expression. RESULTS Protein S significantly reduced ankle joint swelling when overexpressed systemically. Further analysis of knee joints revealed a moderate reduction in pathologic changes in the joint and a significant reduction in the number of splenic Th1 cells when protein S was overexpressed systemically. Local overexpression of GAS-6 decreased joint inflammation and joint pathology. Protein S treatment showed a similar trend of protection. Consistently, GAS-6 and protein S reduced cytokine production in the synovium. Moreover, levels of messenger RNA for interleukin-12 (IL-12) and IL-23 were reduced by GAS-6 and protein S treatment, with a corresponding decrease in the production of interferon-γ and IL-17. TAM ligand overexpression was associated with an increase in SOCS-3 levels, which likely contributed to the amelioration of arthritis. CONCLUSION This study provides the first evidence that TAM receptor stimulation by GAS-6 and protein S can be used to ameliorate arthritis when applied systemically or locally. TAM receptor stimulation limits proinflammatory signaling and adaptive immunity. This pathway provides a novel strategy by which to combat rheumatoid arthritis.
Collapse
Affiliation(s)
- B T van den Brand
- Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Cycloheximide stimulates suppressor of cytokine signaling-3 gene expression in 3T3-L1 adipocytes via the extracellular signal-regulated kinase pathway. Toxicol Lett 2013; 217:42-9. [DOI: 10.1016/j.toxlet.2012.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 11/30/2012] [Accepted: 12/03/2012] [Indexed: 12/20/2022]
|
49
|
Dhar K, Rakesh K, Pankajakshan D, Agrawal DK. SOCS3 promotor hypermethylation and STAT3-NF-κB interaction downregulate SOCS3 expression in human coronary artery smooth muscle cells. Am J Physiol Heart Circ Physiol 2013; 304:H776-85. [PMID: 23335796 DOI: 10.1152/ajpheart.00570.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Suppressor of cytokine signaling-3 (SOCS3) is an intracellular negative regulator of cytokine signaling pathway. We recently found significant reduction in SOCS3 expression in coronary artery smooth muscle cells (CASMCs) of atherosclerotic swine and also in vitro cultured cells. Here, we investigated the underlying mechanisms of SOCS3 downregulation by IGF-1 and TNF-α in human CASMCs(hCASMCs). We propose that hypermethylation of CpG islands in the SOCS3 promoter is responsible for decrease in SOCS3 expression involving STAT3 and NFkB-p65 interaction. Western blot and qPCR data revealed significant upregulation of SOCS3 (6- to 10-fold) in hCASMC when treated individually with TNF-α (100 ng/ml) or IGF-1 (100 ng/ml). However, a significant decrease (5-fold) was observed by the combined treatment with TNF-α and IGF-1 compared with individual stimulation. IGF-1 phosphorylated STAT3 and TNF-α-activated NF-κB in hCASMCs. In the nuclear extract of hCASMCs stimulated with both TNF-α and IGF-1, there was an interaction between NF-κB-p65 and pSTAT3, as determined by co-immunoprecipitation. Knockdown of STAT3 by small interfering RNA abolished SOCS3 expression in response to IGF-1. Methylation-specific PCR confirmed hypermethylation of SOCS3 promoter in hCASMCs stimulated with both TNF-α and IGF-1, and this was positively associated with elevated levels of DNA methyltransferase-I (9- to 10-fold). Knockdown of DNMT1 increased SOCS3 expression in IGF-1+TNF-α-stimulated cells. Downregulation of SOCS3 in the presence of both TNF-α and IGF-1 in hCASMCs is due to SOCS3 promoter hypermethylation involving STAT3-NFkBp65 interaction. Because TNF-α and IGF-1 are released due to mechanical injury during coronary intervention, hypermethylation of SOCS3 gene could be an underlying mechanism of intimal hyperplasia and restenosis.
Collapse
Affiliation(s)
- Kajari Dhar
- Department of Biomedical Sciences, Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA
| | | | | | | |
Collapse
|
50
|
The roles of aerobic exercise training and suppression IL-6 gene expression by RNA interference in the development of insulin resistance. Cytokine 2013; 61:394-405. [PMID: 23294974 DOI: 10.1016/j.cyto.2012.11.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Revised: 10/30/2012] [Accepted: 11/27/2012] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To demonstrate the hypothesis that aerobic exercise training inhibits the development of insulin resistance through IL-6 and probe into the possible molecular mechanism about it. METHODS Rats were raised with high-fat diets for 8 weeks to develop insulin resistance, and glucose infusion rates (GIRs) were determined by hyperinsulinemic-euglycemic clamping to confirm the development of insulin resistance. Aerobic exercise training (the speed and duration time in the first week were respectively 16 m/min and 50 min, and speed increased 1m/min and duration time increased 5 min every week following it) and/or IL-6shRNA plasmid injection (rats received IL-6shRNA injection via the tail vein every two weeks) were adopted during the development of insulin resistance. The serum IL-6, leptin, adiponectin, fasting blood glucose, fasting serum insulin, GIR, IL-6 gene expression levels, p-p38 in various tissues and p-STAT3/t-STAT3 ratio in the liver were measured. RESULTS Rats fed with high-fat diets for 8 weeks were developed insulin resistance and the IL-6mRNA levels of IL-6shRNA injection groups in various tissues were significantly lower than those of control group (P<0.05), respectively. The development of insulin resistance in exercise rats significantly decreased, however, compared with that, the GIR of exercise rats injected by IL-6shRNA was lower (P<0.05). The IL-6mRNA levels were highest in the fat tissue and lowest in the skeletal muscles in all the rats. The serum adiponectin levels decreased (P<0.05) following the development of insulin resistance, and it increased (P<0.05) when the rats were intervened by aerobic exercise training for 8 weeks at the same time. However, there were not significant differences when serum leptin concentrations were compared (P>0.05). The p-p38 significantly increased in the rats fed with high-fat diets, however, p-p38 of the exercise high-fat diets rats in the liver and fat tissues significantly decreased than that (P<0.05). The changes of p-p38 in exercise rats injected by IL-6shRNA were irregular. The activation of STAT3 in the liver significantly increased (P<0.05) following the development of insulin resistance, and it decreased (P<0.05) when the rats were intervened by aerobic exercise training for 8 weeks at the same time, and the gene silencing of IL-6 did not have effects on the activation of STAT3 in the liver (P>0.05). CONCLUSIONS In conclusion, aerobic exercise training prevented the development of insulin resistance through IL-6 to a certain degree. The gene expression and secretion of IL-6 could inhibit the development of insulin resistance. The mechanism of the effects were possibly related with elevating the levels of serum adiponectin, and/or inhibiting the activation of STAT3 in the liver and p38MAPK in the skeletal muscles, liver and fat tissues.
Collapse
|