Cardiotrophin-Like Cytokine Increases Macrophage–Foam Cell Transition

Total Page:16

File Type:pdf, Size:1020Kb

Cardiotrophin-Like Cytokine Increases Macrophage–Foam Cell Transition Cardiotrophin-like Cytokine Increases Macrophage−Foam Cell Transition Sarah Pasquin, Véronique Laplante, Shiriane Kouadri, Andreea Milasan, Gaétan Mayer, Aurélie Jeanne Tormo, This information is current as Virginia Savin, Mukut Sharma, Catherine Martel and of October 7, 2021. Jean-François Gauchat J Immunol published online 12 September 2018 http://www.jimmunol.org/content/early/2018/09/11/jimmun ol.1800733 Downloaded from Why The JI? Submit online. http://www.jimmunol.org/ • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: by guest on October 7, 2021 http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2018 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. Published September 12, 2018, doi:10.4049/jimmunol.1800733 The Journal of Immunology Cardiotrophin-like Cytokine Increases Macrophage–Foam Cell Transition Sarah Pasquin,* Ve´ronique Laplante,* Shiriane Kouadri,* Andreea Milasan,† Gae´tan Mayer,‡ Aure´lie Jeanne Tormo,x Virginia Savin,{ Mukut Sharma,{ Catherine Martel,† and Jean-Franc¸ois Gauchat* CLCF1 is a neurotrophic and B cell–stimulating factor belonging to the IL-6 family. Mutations in the gene coding for CLCF1 or its secretion partner CRLF1 lead to the development of severe phenotypes, suggesting important nonredundant roles in devel- opment, metabolism, and immunity. Although CLCF1 was shown to promote the proliferation of the myeloid cell line M1, its roles on myeloid activation remain underinvestigated. We characterized the effects of CLCF1 on myeloid cells with a focus on monocyte–macrophage and macrophage–foam cell differentiations. CLCF1 injections in mice resulted in a significant increase + in CD11b circulating cells, including proinflammatory monocytes. Furthermore, CLCF1 activated STAT3 phosphorylation in Downloaded from bone marrow CD11b+ cells and in bone marrow–derived macrophages (BMDM). BMDM stimulated with CLCF1 produced a large array of proinflammatory factors comprising IL-6, IL-9, G-CSF, GM-CSF, IL-1b, IL-12, CCL5, and CX3CL1. The pattern of cytokines and chemokines released by CLCF1-treated BMDM led us to investigate the role of CLCF1 in foam cell formation. When pretreated with CLCF1, BMDM presented a marked SR-A1 upregulation, an increase in acetylated–low-density lipopro- tein uptake, and an elevated triglyceride accumulation. CLCF1-induced SR-A1 upregulation, triglyceride accumulation, and acetylated–low-density lipoprotein uptake could be prevented using ruxolitinib, a JAK inhibitor, indicating that the effects of http://www.jimmunol.org/ the cytokine on myeloid cells result from activation of the canonical JAK/STAT signaling pathway. Our data reveal novel biological roles for CLCF1 in the control of myeloid function and identify this cytokine as a strong inducer of macrophage–foam cell transition, thus bringing forward a new potential therapeutic target for atherosclerosis. The Journal of Immunology, 2018, 201: 000–000. therosclerosis is an inflammatory, lipid-driven pathology vascular smooth muscle cells from the media to the fatty lesions, characterized by the development of atherosclerotic contributing to plaque destabilization and vessel disruption (6, 7). plaques in the artery wall (1). When destabilized, these A better understanding of the biological mechanisms underlying A by guest on October 7, 2021 plaques can lead to vessel disruption, causing blood clots and macrophage–foam cell differentiation could lead to the develop- myocardial infarction (2). Atherosclerotic plaques are the result of ment of effective therapies for the treatment of atherosclerosis. lipid-rich cellular accumulation by macrophages, leading to the The gene coding for sortilin, SORT1, has been implicated in formation of foam cells (3–5). Foam cells have a decreased ability low-density lipoprotein (LDL)/cholesterol metabolism, very LDL to migrate and are inefficient at clearing dead cells, causing ne- (VLDL) secretion, and development of atherosclerotic lesions crotic cores in the vessels. Furthermore, foam cells secrete cyto- (8, 9). Sortilin, a ligand for apolipoprotein E (ApoE), LDL, and kines that induce the activation of T cells surrounding the VLDL (10, 11), was recently shown to interact with the composite atherosclerotic lesions. Activated T cells trigger the migration of CLCF1/CRLF1 (12, 13). CLCF1 is an IL-6 family cytokine shown to activate the ciliary neurotrophic factor receptor (CNTFR) (14– 17). Mutations in the gene coding for CLCF1 or its secretion *De´partement de Pharmacologie et Physiologie, Universite´ de Montre´al, Montreal, partner CRLF1 are associated with cold-induced sweating or Quebec H3T 1J4, Canada; †De´partement de Me´decine, Universite´ de Montre´al, Montreal, Quebec H3T 1J4, Canada; ‡Faculte´ de Pharmacie, Universite´ de Montre´al, Crisponi syndrome, which have overlapping phenotypes (18–21). Montreal, Quebec H3T 1J4, Canada; xImmuniT, Montreal, Quebec H2X 1Y4, Canada; Besides dimorphisms, patients tend to suffer from sterile fever { and Renal Division, Kansas City Veterans Affairs Medical Center, Kansas City, episodes in infancy, recurrent infections, life-threatening feeding MO 64128-2226 difficulties, and disinterest in food, suggesting a role for the cy- ORCIDs: 0000-0002-6433-3627 (A.M.); 0000-0003-1328-2169 (A.J.T.). tokine in the regulation of inflammation and metabolism (21). In Received for publication May 24, 2018. Accepted for publication August 16, 2018. mice, transgenic overexpression of CLCF1 led to splenomegaly This work was supported by Canadian Institutes of Health Research Grant MOP- and hypergammaglobulinemia (15). Hemizygous CLCF1 mice 57832 (to J.-F.G.). C.M. is the recipient of a Fonds de Recherche Sante´ Que´bec Research Scholars Career Award Junior 1. present significantly lower circulating leukocytes (http://www. 2/2 Address correspondence and reprint requests to Dr. Jean-Franc¸ois Gauchat, De´parte- mousephenotype.org/), whereas CRLF1 mice (defective for ment de Pharmacologie et Physiologie, Universite´ de Montre´al, Pavillon Roger Gau- CLCF1 secretion) display a strong reduction in bone marrow dry S455, 2900 Edouard Montpetit, Montreal, QC H3T 1J4, Canada. E-mail address: (BM) hematopoietic stem cells (22). Taken together, these obser- [email protected] vations strongly suggest underexplored immunoregulatory roles for Abbreviations used in this article: ac-LDL, acetylated-LDL; ApoE, apolipoprotein E; BM, bone marrow; BMDM, BM-derived macrophage; CNTFR, ciliary neurotrophic CLCF1. factor receptor; CT-1, cardiotrophin-1; iNOS, inducible NO synthase; LDL, low- Recently, we observed that CLCF1 can interact with ApoE, LDL, density lipoprotein; MHC II, MHC class II; SR-A1, scavenger receptor A 1; VLDL, and VLDL and that CLCF1 binding to VLDL modulates the ac- very LDL. tivation of CNTFR, suggesting that CLCF1 is involved in the Copyright Ó 2018 by The American Association of Immunologists, Inc. 0022-1767/18/$35.00 dysregulation of lipid metabolism (23). This led us to investigate www.jimmunol.org/cgi/doi/10.4049/jimmunol.1800733 2 CLCF1 PROMOTES FOAM CELL FORMATION the capability of CLCF1 to activate myeloid cells and induce foam CXNFT; Thermo Fisher Scientific), and 1 mg/test anti–IL-12/IL-23 p40-PE cell differentiation. Results indicate that CLCF1 promotes mac- (clone: C17.8; Thermo Fisher Scientific) in permeabilization buffer for rophage activation, scavenger receptor A 1 (SR-A1) expression, 30 min at room temperature. LDL influx, and foam cell formation. These observations suggest Arginase-1 quantification assay a novel role of CLCF1 in the pathogenesis of atherosclerosis. BMDM were stimulated for 18 h with CLCF1, IFN-g/LPS, or IL-4/IL-13 as described above. Cells were harvested, washed, and lysed for 10 min in Materials and Methods 10 mM Tris-HCL (pH 7.4) containing 1 mM pepstatin A, 1 mM leupeptin, CLCF1 injections in mice and 0.4% (w/v) Triton X-100. Arginase activity was then measured using the Arginase Activity Assay Kit (MAK112; Sigma-Aldrich). Production, purification, and testing of recombinant murine CLCF1 and biotinylated CLCF1 was done as described previously (23). All procedures Cytokines and chemokines quantification conformed to the Canadian Council on Animal Care guidelines and were BMDM were stimulated for 18 h with CLCF1, IFN-g/LPS, or IL-4/IL-13 as approved by the Animal Ethics Committee of the Universite´ de Montre´al. described above. Cell supernatants were collected and analyzed using Eve Six-week-old female C57BL/6 mice were purchased from Charles River Technologies 44-Plex Mouse Cytokines/Chemokines Array (Eve Tech- Laboratories (Saint-Constant, QC). i.p. injections of vehicle (PBS nologies, Calgary, AB). IL-6 production by BMDM was confirmed using 0.1% BSA) or CLCF1 (50 or 300 mg/kg) were administered daily for 5 d, quantitative ELISA according to manufacturer’s instructions (Thermo and mice were euthanized by CO inhalation on day 8. WBC were ana- 2 Fisher Scientific). lyzed
Recommended publications
  • Fog2 Is Critical for Cardiac Function and Maintenance of Coronary Vasculature in the Adult Mouse Heart
    Fog2 is critical for cardiac function and maintenance of coronary vasculature in the adult mouse heart Bin Zhou, … , Sergei G. Tevosian, William T. Pu J Clin Invest. 2009;119(6):1462-1476. https://doi.org/10.1172/JCI38723. Research Article Cardiology Aberrant transcriptional regulation contributes to the pathogenesis of both congenital and adult forms of heart disease. While the transcriptional regulator friend of Gata 2 (FOG2) is known to be essential for heart morphogenesis and coronary development, its tissue-specific function has not been previously investigated. Additionally, little is known about the role of FOG2 in the adult heart. Here we used spatiotemporally regulated inactivation of Fog2 to delineate its function in both the embryonic and adult mouse heart. Early cardiomyocyte-restricted loss of Fog2 recapitulated the cardiac and coronary defects of the Fog2 germline murine knockouts. Later cardiomyocyte-restricted loss ofF og2 (Fog2MC) did not result in defects in cardiac structure or coronary vessel formation. However, Fog2MC adult mice had severely depressed ventricular function and died at 8–14 weeks. Fog2MC adult hearts displayed a paucity of coronary vessels, associated with myocardial hypoxia, increased cardiomyocyte apoptosis, and cardiac fibrosis. Induced inactivation of Fog2 in the adult mouse heart resulted in similar phenotypes, as did ablation of the FOG2 interaction with the transcription factor GATA4. Loss of the FOG2 or FOG2-GATA4 interaction altered the expression of a panel of angiogenesis-related genes. Collectively, our data indicate that FOG2 regulates adult heart function and coronary angiogenesis. Find the latest version: https://jci.me/38723/pdf Research article Fog2 is critical for cardiac function and maintenance of coronary vasculature in the adult mouse heart Bin Zhou,1,2 Qing Ma,1 Sek Won Kong,1 Yongwu Hu,1,3 Patrick H.
    [Show full text]
  • Cytokine Nomenclature
    RayBiotech, Inc. The protein array pioneer company Cytokine Nomenclature Cytokine Name Official Full Name Genbank Related Names Symbol 4-1BB TNFRSF Tumor necrosis factor NP_001552 CD137, ILA, 4-1BB ligand receptor 9 receptor superfamily .2. member 9 6Ckine CCL21 6-Cysteine Chemokine NM_002989 Small-inducible cytokine A21, Beta chemokine exodus-2, Secondary lymphoid-tissue chemokine, SLC, SCYA21 ACE ACE Angiotensin-converting NP_000780 CD143, DCP, DCP1 enzyme .1. NP_690043 .1. ACE-2 ACE2 Angiotensin-converting NP_068576 ACE-related carboxypeptidase, enzyme 2 .1 Angiotensin-converting enzyme homolog ACTH ACTH Adrenocorticotropic NP_000930 POMC, Pro-opiomelanocortin, hormone .1. Corticotropin-lipotropin, NPP, NP_001030 Melanotropin gamma, Gamma- 333.1 MSH, Potential peptide, Corticotropin, Melanotropin alpha, Alpha-MSH, Corticotropin-like intermediary peptide, CLIP, Lipotropin beta, Beta-LPH, Lipotropin gamma, Gamma-LPH, Melanotropin beta, Beta-MSH, Beta-endorphin, Met-enkephalin ACTHR ACTHR Adrenocorticotropic NP_000520 Melanocortin receptor 2, MC2-R hormone receptor .1 Activin A INHBA Activin A NM_002192 Activin beta-A chain, Erythroid differentiation protein, EDF, INHBA Activin B INHBB Activin B NM_002193 Inhibin beta B chain, Activin beta-B chain Activin C INHBC Activin C NM005538 Inhibin, beta C Activin RIA ACVR1 Activin receptor type-1 NM_001105 Activin receptor type I, ACTR-I, Serine/threonine-protein kinase receptor R1, SKR1, Activin receptor-like kinase 2, ALK-2, TGF-B superfamily receptor type I, TSR-I, ACVRLK2 Activin RIB ACVR1B
    [Show full text]
  • Despite Differential Gene Expression Profiles Pediatric MDS Derived Mesenchymal Stromal Cells Display Functionality in Vitro☆ F.G.J
    Stem Cell Research (2015) 14, 198-210 Available online at www.sciencedirect.com ScienceDirect www.elsevier.com/locate/scr Despite differential gene expression profiles pediatric MDS derived mesenchymal stromal cells display functionality in vitro☆ F.G.J. Calkoen a,⁎, C. Vervat a, M. van Pel b, V. de Haas c, L.S. Vijfhuizen d, E. Eising d, W.G.M. Kroes e, P.A.C. 't Hoen d, M.M. van den Heuvel-Eibrink c,f, R.M. Egeler a,g, M.J.D. van Tol a, L.M. Ball a a Department of Pediatrics, Section Immunology, Hematology/Oncology and Hematopoietic Stem Cell Transplantation, Leiden University Medical Center, Leiden, The Netherlands b Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands c Dutch Childhood Oncology Group (DCOG), The Hague, The Netherlands d Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands e Laboratory for Diagnostic Genome Analysis, Leiden University Medical Center, Leiden, The Netherlands f Department of Pediatric Oncology/Hematology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands g Department of Hematology/Oncology and Hematopoietic Stem Cell Transplantation, Hospital for Sick Children, University of Toronto, Toronto, Canada Received 24 June 2014; received in revised form 3 December 2014; accepted 19 January 2015 Available online 28 January 2015 Abstract Pediatric myelodysplastic syndrome (MDS) is a heterogeneous disease covering a spectrum ranging from aplasia (RCC) to myeloproliferation (RAEB(t)). In adult-type MDS there is increasing evidence for abnormal function of the bone-marrow microenvironment. Here, we extensively studied the mesenchymal stromal cells (MSCs) derived from children with MDS.
    [Show full text]
  • Supplemental Materials ZNF281 Enhances Cardiac Reprogramming
    Supplemental Materials ZNF281 enhances cardiac reprogramming by modulating cardiac and inflammatory gene expression Huanyu Zhou, Maria Gabriela Morales, Hisayuki Hashimoto, Matthew E. Dickson, Kunhua Song, Wenduo Ye, Min S. Kim, Hanspeter Niederstrasser, Zhaoning Wang, Beibei Chen, Bruce A. Posner, Rhonda Bassel-Duby and Eric N. Olson Supplemental Table 1; related to Figure 1. Supplemental Table 2; related to Figure 1. Supplemental Table 3; related to the “quantitative mRNA measurement” in Materials and Methods section. Supplemental Table 4; related to the “ChIP-seq, gene ontology and pathway analysis” and “RNA-seq” and gene ontology analysis” in Materials and Methods section. Supplemental Figure S1; related to Figure 1. Supplemental Figure S2; related to Figure 2. Supplemental Figure S3; related to Figure 3. Supplemental Figure S4; related to Figure 4. Supplemental Figure S5; related to Figure 6. Supplemental Table S1. Genes included in human retroviral ORF cDNA library. Gene Gene Gene Gene Gene Gene Gene Gene Symbol Symbol Symbol Symbol Symbol Symbol Symbol Symbol AATF BMP8A CEBPE CTNNB1 ESR2 GDF3 HOXA5 IL17D ADIPOQ BRPF1 CEBPG CUX1 ESRRA GDF6 HOXA6 IL17F ADNP BRPF3 CERS1 CX3CL1 ETS1 GIN1 HOXA7 IL18 AEBP1 BUD31 CERS2 CXCL10 ETS2 GLIS3 HOXB1 IL19 AFF4 C17ORF77 CERS4 CXCL11 ETV3 GMEB1 HOXB13 IL1A AHR C1QTNF4 CFL2 CXCL12 ETV7 GPBP1 HOXB5 IL1B AIMP1 C21ORF66 CHIA CXCL13 FAM3B GPER HOXB6 IL1F3 ALS2CR8 CBFA2T2 CIR1 CXCL14 FAM3D GPI HOXB7 IL1F5 ALX1 CBFA2T3 CITED1 CXCL16 FASLG GREM1 HOXB9 IL1F6 ARGFX CBFB CITED2 CXCL3 FBLN1 GREM2 HOXC4 IL1F7
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • GP130 Cytokines in Breast Cancer and Bone
    cancers Review GP130 Cytokines in Breast Cancer and Bone Tolu Omokehinde 1,2 and Rachelle W. Johnson 1,2,3,* 1 Program in Cancer Biology, Vanderbilt University, Nashville, TN 37232, USA; [email protected] 2 Vanderbilt Center for Bone Biology, Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA 3 Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA * Correspondence: [email protected]; Tel.: +1-615-875-8965 Received: 14 December 2019; Accepted: 29 January 2020; Published: 31 January 2020 Abstract: Breast cancer cells have a high predilection for skeletal homing, where they may either induce osteolytic bone destruction or enter a latency period in which they remain quiescent. Breast cancer cells produce and encounter autocrine and paracrine cytokine signals in the bone microenvironment, which can influence their behavior in multiple ways. For example, these signals can promote the survival and dormancy of bone-disseminated cancer cells or stimulate proliferation. The interleukin-6 (IL-6) cytokine family, defined by its use of the glycoprotein 130 (gp130) co-receptor, includes interleukin-11 (IL-11), leukemia inhibitory factor (LIF), oncostatin M (OSM), ciliary neurotrophic factor (CNTF), and cardiotrophin-1 (CT-1), among others. These cytokines are known to have overlapping pleiotropic functions in different cell types and are important for cross-talk between bone-resident cells. IL-6 cytokines have also been implicated in the progression and metastasis of breast, prostate, lung, and cervical cancer, highlighting the importance of these cytokines in the tumor–bone microenvironment. This review will describe the role of these cytokines in skeletal remodeling and cancer progression both within and outside of the bone microenvironment.
    [Show full text]
  • Early Growth Response 1 Regulates Hematopoietic Support and Proliferation in Human Primary Bone Marrow Stromal Cells
    Hematopoiesis SUPPLEMENTARY APPENDIX Early growth response 1 regulates hematopoietic support and proliferation in human primary bone marrow stromal cells Hongzhe Li, 1,2 Hooi-Ching Lim, 1,2 Dimitra Zacharaki, 1,2 Xiaojie Xian, 2,3 Keane J.G. Kenswil, 4 Sandro Bräunig, 1,2 Marc H.G.P. Raaijmakers, 4 Niels-Bjarne Woods, 2,3 Jenny Hansson, 1,2 and Stefan Scheding 1,2,5 1Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, Lund, Sweden; 2Lund Stem Cell Center, Depart - ment of Laboratory Medicine, Lund University, Lund, Sweden; 3Division of Molecular Medicine and Gene Therapy, Department of Labora - tory Medicine, Lund University, Lund, Sweden; 4Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands and 5Department of Hematology, Skåne University Hospital Lund, Skåne, Sweden ©2020 Ferrata Storti Foundation. This is an open-access paper. doi:10.3324/haematol. 2019.216648 Received: January 14, 2019. Accepted: July 19, 2019. Pre-published: August 1, 2019. Correspondence: STEFAN SCHEDING - [email protected] Li et al.: Supplemental data 1. Supplemental Materials and Methods BM-MNC isolation Bone marrow mononuclear cells (BM-MNC) from BM aspiration samples were isolated by density gradient centrifugation (LSM 1077 Lymphocyte, PAA, Pasching, Austria) either with or without prior incubation with RosetteSep Human Mesenchymal Stem Cell Enrichment Cocktail (STEMCELL Technologies, Vancouver, Canada) for lineage depletion (CD3, CD14, CD19, CD38, CD66b, glycophorin A). BM-MNCs from fetal long bones and adult hip bones were isolated as reported previously 1 by gently crushing bones (femora, tibiae, fibulae, humeri, radii and ulna) in PBS+0.5% FCS subsequent passing of the cell suspension through a 40-µm filter.
    [Show full text]
  • CRLF1 Promotes Malignant Phenotypes of Papillary Thyroid
    Yu et al. Cell Death and Disease (2018) 9:371 DOI 10.1038/s41419-018-0352-0 Cell Death & Disease ARTICLE Open Access CRLF1 promotes malignant phenotypes of papillary thyroid carcinoma by activating the MAPK/ERK and PI3K/AKT pathways Shi-Tong Yu1,2, Qian Zhong3,Ren-HuiChen1,2,PingHan1,2,Shi-BingLi3,HuaZhang3,LiYuan3, Tian-Liang Xia3, Mu-Sheng Zeng 3 and Xiao-Ming Huang1,2 Abstract Papillary thyroid carcinoma (PTC) is the one of the most common types of endocrine cancer and has a heterogeneous prognosis. Tumors from patients with poor prognosis may differentially express specific genes. Therefore, an analysis of The Cancer Genome Atlas (TCGA) database was performed and revealed that cytokine receptor-like factor 1 (CRLF1) may be a potential novel target for PTC treatment. The objective of the current study was to explore the expression of CRLF1 in PTC and to investigate the main functions and mechanisms of CRLF1 in PTC. PTC tissues exhibited higher CRLF1 expression at both the mRNA and protein levels than it did with normal thyroid tissues. High CRLF1 levels were associated with aggressive clinicopathological features and poor disease-free survival rates. By using loss-of-function and gain-of-function assays, we found that CRLF1 not only increased cell migration and invasion in vitro but also promoted tumor growth both in vitro and in vivo. In addition, CRLF1 induced epithelial–mesenchymal transitions. Overexpression of CRLF1 activated the ERK1/2 and AKT pathways. The oncogenic effects induced by CRLF1 were suppressed by treating the cells with the MEK inhibitor U0126 or the AKT inhibitor MK-2206.
    [Show full text]
  • Impacts of Ciliary Neurotrophic Factor on the Retinal Transcriptome in a Mouse Model of Photoreceptor Degeneration
    www.nature.com/scientificreports OPEN Impacts of ciliary neurotrophic factor on the retinal transcriptome in a mouse model of photoreceptor degeneration Yanjie Wang1, Kun-Do Rhee1, Matteo Pellegrini2 & Xian-Jie Yang1* Ciliary neurotrophic factor (CNTF) has been tested in clinical trials for human retinal degeneration due to its potent neuroprotective efects in various animal models. To decipher CNTF-triggered molecular events in the degenerating retina, we performed high-throughput RNA sequencing analyses using the Rds/Prph2 (P216L) transgenic mouse as a preclinical model for retinitis pigmentosa. In the absence of CNTF treatment, transcriptome alterations were detected at the onset of rod degeneration compared with wild type mice, including reduction of key photoreceptor transcription factors Crx, Nrl, and rod phototransduction genes. Short-term CNTF treatments caused further declines of photoreceptor transcription factors accompanied by marked decreases of both rod- and cone-specifc gene expression. In addition, CNTF triggered acute elevation of transcripts in the innate immune system and growth factor signaling. These immune responses were sustained after long-term CNTF exposures that also afected neuronal transmission and metabolism. Comparisons of transcriptomes also uncovered common pathways shared with other retinal degeneration models. Cross referencing bulk RNA-seq with single-cell RNA-seq data revealed the CNTF responsive cell types, including Müller glia, rod and cone photoreceptors, and bipolar cells. Together, these results demonstrate the infuence of exogenous CNTF on the retinal transcriptome landscape and illuminate likely CNTF impacts in degenerating human retinas. Retinal degeneration (RD) is known as an irreversible, progressive neurologic disorder caused by genetic muta- tions and/or environmental or pathological damage to the retina.
    [Show full text]
  • Association of Cardiotrophin-Like Cytokine Factor 1 Levels in Peripheral
    Chen et al. BMC Musculoskeletal Disorders (2021) 22:62 https://doi.org/10.1186/s12891-020-03924-9 RESEARCH ARTICLE Open Access Association of cardiotrophin-like cytokine factor 1 levels in peripheral blood mononuclear cells with bone mineral density and osteoporosis in postmenopausal women Xuan Chen1†, Jianyang Li2†, Yunjin Ye1, Jingwen Huang1, Lihua Xie1, Juan Chen1, Shengqiang Li1, Sainan Chen1 and Jirong Ge1* Abstract Background: Recent research has suggested that cardiotrophin-like cytokine factor 1 (CLCF1) may be an important regulator of bone homeostasis. Furthermore, a whole gene chip analysis suggested that the expression levels of CLCF1 in the peripheral blood mononuclear cells (PBMCs) were downregulated in postmenopausal women with osteoporosis. This study aimed to assess whether the expression levels of CLCF1 in PBMCs can reflect the severity of bone mass loss and the related fracture risk. Methods: In all, 360 postmenopausal women, aged 50 to 80 years, were included in the study. A survey to evaluate the participants’ health status, measurement of bone mineral density (BMD), routine blood test, and CLCF1 expression level test were performed. Results: Based on the participants’ bone health, 27 (7.5%), 165 (45.83%), and 168 (46.67%) participants were divided into the normal, osteopenia, and osteoporosis groups, respectively. CLCF1 protein levels in the normal and osteopenia groups were higher than those in the osteoporosis group. While the CLCF1 mRNA level was positively associated with the BMD of total femur (r =0.169,p = 0.011) and lumbar spine (r =0.176,p = 0.001), the protein level was positively associated with the BMD of the lumbar spine (r =0.261,p <0.001),femoralneck(r =0.236,p = 0.001), greater trochanter (r =0.228,p =0.001),andWard’striangle(r =0.149,p = 0.036).
    [Show full text]
  • Engineering a Potent Receptor Superagonist Or Antagonist from a Novel IL-6 Family Cytokine Ligand
    Engineering a potent receptor superagonist or antagonist from a novel IL-6 family cytokine ligand Jun W. Kima, Cesar P. Marquezb,c, R. Andres Parra Sperberga, Jiaxiang Wua,d, Won G. Baee, Po-Ssu Huanga, E. Alejandro Sweet-Corderob,1, and Jennifer R. Cochrana,f,1 aDepartment of Bioengineering, Stanford University, Stanford, CA 94305; bDivision of Hematology and Oncology, Department of Pediatrics, University of California, San Francisco, CA 94158; cSchool of Medicine, Stanford University, Stanford, CA 94305; dTencent AI Lab, 518000 Shenzhen, China; eDepartment of Electrical Engineering, Soongsil University, 156-743 Seoul, Korea; and fDepartment of Chemical Engineering, Stanford University, Stanford, CA 94305 Edited by Joseph Schlessinger, Yale University, New Haven, CT, and approved May 5, 2020 (received for review December 26, 2019) Interleukin-6 (IL-6) family cytokines signal through multimeric re- facilitate neuronal regeneration, while a CNTFR antagonist could ceptor complexes, providing unique opportunities to create novel inhibit this signaling axis for cancer or other disease treatment. ligand-based therapeutics. The cardiotrophin-like cytokine factor 1 We used a combinatorial screening approach facilitated by yeast (CLCF1) ligand has been shown to play a role in cancer, osteopo- surface display to identify CLCF1 variants that altered receptor- rosis, and atherosclerosis. Once bound to ciliary neurotrophic fac- mediated cell signaling and biochemical function in disparate tor receptor (CNTFR), CLCF1 mediates interactions to coreceptors ways. CLCF1 variants with significantly increased CNTFR affinity glycoprotein 130 (gp130) and leukemia inhibitory factor receptor drove enhanced tripartite receptor complex formation and func- (LIFR). By increasing CNTFR-mediated binding to these coreceptors tioned as superagonists of cell signaling and axon regeneration.
    [Show full text]
  • Development and Validation of a Protein-Based Risk Score for Cardiovascular Outcomes Among Patients with Stable Coronary Heart Disease
    Supplementary Online Content Ganz P, Heidecker B, Hveem K, et al. Development and validation of a protein-based risk score for cardiovascular outcomes among patients with stable coronary heart disease. JAMA. doi: 10.1001/jama.2016.5951 eTable 1. List of 1130 Proteins Measured by Somalogic’s Modified Aptamer-Based Proteomic Assay eTable 2. Coefficients for Weibull Recalibration Model Applied to 9-Protein Model eFigure 1. Median Protein Levels in Derivation and Validation Cohort eTable 3. Coefficients for the Recalibration Model Applied to Refit Framingham eFigure 2. Calibration Plots for the Refit Framingham Model eTable 4. List of 200 Proteins Associated With the Risk of MI, Stroke, Heart Failure, and Death eFigure 3. Hazard Ratios of Lasso Selected Proteins for Primary End Point of MI, Stroke, Heart Failure, and Death eFigure 4. 9-Protein Prognostic Model Hazard Ratios Adjusted for Framingham Variables eFigure 5. 9-Protein Risk Scores by Event Type This supplementary material has been provided by the authors to give readers additional information about their work. Downloaded From: https://jamanetwork.com/ on 10/02/2021 Supplemental Material Table of Contents 1 Study Design and Data Processing ......................................................................................................... 3 2 Table of 1130 Proteins Measured .......................................................................................................... 4 3 Variable Selection and Statistical Modeling ........................................................................................
    [Show full text]