301
|
Bazzi S, El-Darzi E, McDowell T, Modjtahedi H, Mudan S, Achkar M, Akle C, Kadara H, Bahr GM. Defining Genome-Wide Expression and Phenotypic Contextual Cues in Macrophages Generated by Granulocyte/Macrophage Colony-Stimulating Factor, Macrophage Colony-Stimulating Factor, and Heat-Killed Mycobacteria. Front Immunol 2017; 8:1253. [PMID: 29046677 PMCID: PMC5632758 DOI: 10.3389/fimmu.2017.01253] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 09/20/2017] [Indexed: 12/23/2022] Open
Abstract
Heat-killed (HK) Mycobacterium obuense (NCTC13365) is currently being evaluated in the clinic as an immunotherapeutic agent for cancer treatment. Yet, the molecular underpinnings underlying immunomodulatory properties of HK M. obuense are still largely undefined. To fill this void, we sought to perform immunophenotyping, chemokine/cytokine release analysis and genome-wide characterization of monocyte-derived macrophages (MDM) in which monocytes were originally isolated from healthy donors and differentiated by HK M. obuense (Mob-MDM) relative to macrophage colony-stimulating factor (M-MDM) and granulocyte/macrophage colony-stimulating factor (GM-MDM). Immunophenotyping and cytokine release analysis revealed downregulated surface expression of CD36, decreased spontaneous release of CCL2 and increased spontaneous secretion of CCL5, CXCL8/IL-8, IL-6, and TNF-α in Mob-MDM relative to M-MDM and GM-MDM. Analysis of cytostatic activity showed that Mob-MDM exhibited similar growth inhibitory effects on immortalized and malignant epithelial cells compared with GM-MDM but at an elevated rate relative to M-MDM. To understand global cues in Mob-MDM, we performed comparative RNA-sequencing (RNA-Seq) analysis of Mob-MDM relative to GM-MDM and M-MDM (n = 4 donors). Clustering analysis underscored expression profiles (n = 256) that were significantly modulated in Mob-MDM versus both M-MDM and GM-MDM including, among others, chemokines/cytokines and their receptors, enzymes and transcriptions factors. Topological functional analysis of these profiles identified pathways and gene sets linked to Mob-MDM phenotype including nitric oxide production, acute phase response signaling and microbe recognition pathways as well as signaling cues mediated by the proinflammatory cytokine, interferon-gamma, and the intracellular pattern recognition receptor, nucleotide-binding oligomerization domain-containing protein 2. Taken together, our study highlights molecular immune phenotypes and global signaling cues in Mob-MDM that may underlie immunomodulatory properties of HK M. obuense. Such properties could be of valuable use in immunotherapy approaches such as adoptive cell therapy against cancer.
Collapse
Affiliation(s)
- Samer Bazzi
- Faculty of Science, Engineering and Computing, School of Life Sciences, Kingston University, Kingston upon Thames, United Kingdom.,Faculty of Sciences, University of Balamand, Al Kurah, Lebanon
| | - Emale El-Darzi
- Faculty of Medicine and Medical Sciences, University of Balamand, Al Kurah, Lebanon
| | - Tina McDowell
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Helmout Modjtahedi
- Faculty of Science, Engineering and Computing, School of Life Sciences, Kingston University, Kingston upon Thames, United Kingdom
| | - Satvinder Mudan
- St George's University of London, Imperial College, London and The Royal Marsden Hospital, London, United Kingdom
| | - Marcel Achkar
- Clinical Laboratory, Nini Hospital, Tripoli, Lebanon
| | - Charles Akle
- Immodulon Therapeutics Ltd., Uxbridge, United Kingdom
| | - Humam Kadara
- Faculty of Medicine, Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Georges M Bahr
- Faculty of Medicine and Medical Sciences, University of Balamand, Al Kurah, Lebanon
| |
Collapse
|
302
|
López-Cotarelo P, Gómez-Moreira C, Criado-García O, Sánchez L, Rodríguez-Fernández JL. Beyond Chemoattraction: Multifunctionality of Chemokine Receptors in Leukocytes. Trends Immunol 2017; 38:927-941. [PMID: 28935522 DOI: 10.1016/j.it.2017.08.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 06/05/2017] [Accepted: 08/08/2017] [Indexed: 12/19/2022]
Abstract
The word chemokine is a combination of the words chemotactic and cytokine, in other words cytokines that promote chemotaxis. Hence, the term chemokine receptor refers largely to the ability to regulate chemoattraction. However, these receptors can modulate additional leukocyte functions, as exemplified by the case of CCR7 which, apart from chemotaxis, regulates survival, migratory speed, endocytosis, differentiation and cytoarchitecture. We present evidence highlighting that multifunctionality is a common feature of chemokine receptors. Based on the activities that they regulate, we suggest that chemokine receptors can be classified into inflammatory (which control both inflammatory and homeostatic functions) and homeostatic families. The information accrued also suggests that the non-chemotactic functions controlled by chemokine receptors may contribute to optimizing leukocyte functioning under normal physiological conditions and during inflammation.
Collapse
Affiliation(s)
- Pilar López-Cotarelo
- Molecular Microbiology and Infection Biology Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain; Equal first authors
| | - Carolina Gómez-Moreira
- Molecular Microbiology and Infection Biology Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain; Equal first authors
| | - Olga Criado-García
- Molecular Microbiology and Infection Biology Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain; Equal first authors
| | - Lucas Sánchez
- Cellular and Molecular Biology Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - José Luis Rodríguez-Fernández
- Molecular Microbiology and Infection Biology Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain.
| |
Collapse
|
303
|
Xu X, Guo Y, Zhao J, He S, Wang Y, Lin Y, Wang N, Liu Q. Punicalagin, a PTP1B inhibitor, induces M2c phenotype polarization via up-regulation of HO-1 in murine macrophages. Free Radic Biol Med 2017; 110:408-420. [PMID: 28690198 DOI: 10.1016/j.freeradbiomed.2017.06.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 05/26/2017] [Accepted: 06/17/2017] [Indexed: 11/28/2022]
Abstract
Current data have shown that punicalagin (PUN), an ellagitannin isolated from pomegranate, possesses anti-inflammatory and anti-oxidant properties; however, its direct targets have not yet been reported. This is the first report that PTP1B serves as a direct target of PUN, with IC50 value of 1.04μM. Results from NPOI further showed that the Kon and Koff of PUN-PTP1B complex were 3.38e2M-1s-1 and 4.13e-3s-1, respectively. The active site Arg24 of PTP1B was identified as a key binding site of PUN by computation simulation and point mutation. Moreover, inhibition of PTP1B by PUN promoted an M2c-like macrophage polarization and enhanced anti-inflammatory cytokines expression, including IL-10 and M-CSF. Based on gene expression profile, we elucidated that PUN treatment significantly up-regulated 275 genes and down-regulated 1059 genes. M1-like macrophage marker genes, such as Tlr4, Irf1/2, Hmgb1, and Stat1 were down-regulated, while M2 marker genes, including Tmem171, Gpr35, Csf1, Il1rn, Cebpb, Fos, Vegfα, Slc11a1, and Bhlhe40 were up-regulated in PUN-treated macrophages. Hmox-1, a gene encoding HO-1 protein, was preferentially expressed with 16-fold change. Inhibition of HO-1 obviously restored PUN-induced M2 polarization and IL-10 secretion. In addition, phosphorylation of both Akt and STAT3 contributed to PUN-induced HO-1 expression. This study provided new insights into the mechanisms of PUN-mediated anti-inflammatory and anti-oxidant activities and provided new therapeutic strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Xiaolong Xu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China; Beijing Institute of Traditional Chinese Medicine, Beijing 100010, PR China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, PR China
| | - Yuhong Guo
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China; Beijing Institute of Traditional Chinese Medicine, Beijing 100010, PR China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, PR China
| | - Jingxia Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China; Beijing Institute of Traditional Chinese Medicine, Beijing 100010, PR China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, PR China
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China; Beijing Institute of Traditional Chinese Medicine, Beijing 100010, PR China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, PR China
| | - Yan Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China; Beijing Institute of Traditional Chinese Medicine, Beijing 100010, PR China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, PR China
| | - Yan Lin
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China; Beijing Institute of Traditional Chinese Medicine, Beijing 100010, PR China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, PR China
| | - Ning Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China; Beijing Institute of Traditional Chinese Medicine, Beijing 100010, PR China
| | - Qingquan Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, PR China; Beijing Institute of Traditional Chinese Medicine, Beijing 100010, PR China; Beijing Key Laboratory of Basic Research with Traditional Chinese Medicine on Infectious Diseases, Beijing 100010, PR China.
| |
Collapse
|
304
|
Boro M, Balaji KN. CXCL1 and CXCL2 Regulate NLRP3 Inflammasome Activation via G-Protein-Coupled Receptor CXCR2. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1660-1671. [PMID: 28739876 DOI: 10.4049/jimmunol.1700129] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 06/21/2017] [Indexed: 01/03/2025]
Abstract
Inflammation is an extensively concerted process that confers protection to the host encountering immune insult. The major inflammatory mediators include IL-1 family members, such as IL-1β, and the functional activation of such molecules is arbitrated by their regulated cleavage brought about by components of a multiprotein complex called inflammasome. In this context, NLR family pyrin domain containing 3 (NLRP3) inflammasome activation often acts as a rate-limiting step in regulating critical cell-fate decisions in various inflammatory scenarios. In this study, we identify the G-protein-coupled receptor CXCR2 (recognizing chemokines CXCL1 and CXCL2) as another arm feeding into the regulated activation of NLRP3 inflammasome in macrophages. We demonstrate that in vivo blocking of CXCL1 and CXCL2 can significantly reduce the Mycobacterium tuberculosis-induced bioactive IL-1β production. Further, CXCL1 could amplify the inflammasome activation in in vivo mouse models of carrageenan-induced inflammation in footpads and air pouches. The mechanistic insights revealed CXCR2-driven protein kinase C μ-dependent integrin-linked kinase to be essential for CXCL1-mediated activation of NLRP3 inflammasome. Blocking the activity of integrin-linked kinase or protein kinase C μ either by small interfering RNA-mediated knockdown or pharmacological inhibitor compromised inflammasome activation and subsequent production of bioactive IL-1β. Taken together, our study demonstrates CXCR2-driven activation of NLRP3 inflammasome in macrophages and indicates a potential host-directed therapeutic target to limit the damaging inflammation associated with overt production of proinflammatory IL-1β.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/administration & dosage
- Cells, Cultured
- Chemokine CXCL1/immunology
- Chemokine CXCL1/metabolism
- Chemokine CXCL2/immunology
- Chemokine CXCL2/metabolism
- Humans
- Inflammasomes/metabolism
- Interleukin-1beta/immunology
- Macrophages/immunology
- Mice
- Mice, Inbred BALB C
- Mycobacterium tuberculosis/immunology
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- RNA, Small Interfering/genetics
- Receptors, G-Protein-Coupled/immunology
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Interleukin-8B/immunology
- Receptors, Interleukin-8B/metabolism
- Tuberculosis/immunology
Collapse
Affiliation(s)
- Monoranjan Boro
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
305
|
SOCS molecules: the growing players in macrophage polarization and function. Oncotarget 2017; 8:60710-60722. [PMID: 28948005 PMCID: PMC5601173 DOI: 10.18632/oncotarget.19940] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023] Open
Abstract
The concept of macrophage polarization is defined in terms of macrophage phenotypic heterogeneity and functional diversity. Cytokines signals are thought to be required for the polarization of macrophage populations toward different phenotypes at different stages in development, homeostasis and disease. The suppressors of cytokine signaling family of proteins contribute to the magnitude and duration of cytokines signaling, which ultimately control the subtle adjustment of the balance between divergent macrophage phenotypes. This review highlights the specific roles and mechanisms of various cytokines family and their negative regulators link to the macrophage polarization programs. Eventually, breakthrough in the identification of these molecules will provide the novel therapeutic approaches for a host of diseases by targeting macrophage phenotypic shift.
Collapse
|
306
|
Nakata R, Shimada H, Fernandez GE, Fanter R, Fabbri M, Malvar J, Zimmermann P, DeClerck YA. Contribution of neuroblastoma-derived exosomes to the production of pro-tumorigenic signals by bone marrow mesenchymal stromal cells. J Extracell Vesicles 2017; 6:1332941. [PMID: 28717423 PMCID: PMC5505006 DOI: 10.1080/20013078.2017.1332941] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Indexed: 12/20/2022] Open
Abstract
The bone marrow (BM) niche is a microenvironment promoting survival, dormancy and therapeutic resistance in tumor cells. Central to this function are mesenchymal stromal cells (MSCs). Here, using neuroblastoma (NB) as a model, we demonstrate that NB cells release an extracellular vesicle (EVs) whose protein cargo is enriched in exosomal proteins but lacks cytokines and chemokines. Using three different purification methods, we then demonstrate that NB-derived exosomes were captured by MSCs and induced the production of pro-tumorigenic cytokines and chemokines, including interleukin-6 (IL-6), IL-8/CXCL8, vascular endothelial cell growth factor and monocyte-chemotactic protein-1, with exosomes prepared by size exclusion chromatography having the highest activity. We found no correlation between the IL-6 and IL-8/CXCL8 stimulatory activity of exosomes from eight NB cell lines and their origin, degree of MYCN amplification, drug resistance and disease status. We then demonstrate that the uptake of NB exosomes by MSCs was associated with a rapid increase in ERK1/2 and AKT activation, and that blocking ERK1/2 but not AKT activation inhibited the IL-6 and IL-8/CXCL8 production by MSCs without affecting exosome uptake. Thus, we describe a new mechanism by which NB cells induce in MSCs an inflammatory reaction that contributes to a favorable microenvironment in the BM.
Collapse
Affiliation(s)
- Rie Nakata
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Southern California, Los Angeles, CA, USA.,Department of Pediatrics, University of Southern California, Los Angeles, CA, USA
| | - Hiroyuki Shimada
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Department of Pathology and Laboratory Medicine, University of Southern California, Los Angeles, CA, USA
| | - G Esteban Fernandez
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Rob Fanter
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA
| | - Muller Fabbri
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Southern California, Los Angeles, CA, USA.,Department of Pediatrics, University of Southern California, Los Angeles, CA, USA.,Department of Molecular Microbiology and Immunology, University of Southern California, Los Angeles, CA, USA
| | - Jemily Malvar
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Southern California, Los Angeles, CA, USA
| | - Pascale Zimmermann
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068-CNRS UMR7258, Aix-Marseille Université, Institut Paoli-Calmettes, Marseille, France
| | - Yves A DeClerck
- The Saban Research Institute of Children's Hospital Los Angeles, University of Southern California, Los Angeles, CA, USA.,Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Southern California, Los Angeles, CA, USA.,Department of Pediatrics, University of Southern California, Los Angeles, CA, USA.,Department of Biochemistry and Molecular Biology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
307
|
McGrath-Morrow SA, Ndeh R, Collaco JM, Poupore AK, Dikeman D, Zhong Q, Singer BD, D'Alessio F, Scott A. The innate immune response to lower respiratory tract E. Coli infection and the role of the CCL2-CCR2 axis in neonatal mice. Cytokine 2017. [PMID: 28628889 DOI: 10.1016/j.cyto.2017.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Neonates have greater morbidity/mortality from lower respiratory tract infections (LRTI) compared to older children. Lack of conditioning of the pulmonary immune system due to limited environmental exposures and/or infectious challenges likely contributes to the increase susceptibility in the neonate. In this study, we sought to gain insights into the nature and dynamics of the neonatal pulmonary immune response to LRTI using a murine model. METHODS Wildtype (WT) and Ccr2-/- C57BL/6 neonatal and juvenile mice received E. coli or PBS by direct pharyngeal aspiration. Flow cytometry was used to measure immune cell dynamics and identify cytokine-producing cells. Real-time PCR and ELISA were used to measure cytokine/chemokine expression. RESULTS Innate immune cell recruitment in response to E. coli-induced LRTI was delayed in the neonatal lung compared to juvenile lung. Lung clearance of bacteria was also significantly delayed in the neonate. Ccr2-/- neonates, which lack an intact CCL2-CCR2 axis, had higher mortality after E. coli challenged than Ccr2+/+ neonates. A greater percentage of CD8+ T cells and monocytes from WT neonates challenged with E. coli produced TNF compared to controls. CONCLUSION The pulmonary immune response to E. coli-induced LRTI differed significantly between neonatal and juvenile mice. Neonates were more susceptible to increasing doses of E. coli and exhibited greater mortality than juveniles. In the absence of an intact CCL2-CCR2 axis, susceptibility to LRTI-induced mortality was further increased in neonatal mice. Taken together these findings underscore the importance of age-related differences in the innate immune response to LRTI during early stages of postnatal life.
Collapse
Affiliation(s)
- Sharon A McGrath-Morrow
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States.
| | - Roland Ndeh
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Joseph M Collaco
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Amy K Poupore
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Baltimore, MD, United States
| | - Dustin Dikeman
- Eudowood Division of Pediatric Respiratory Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Qiong Zhong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, United States
| | - Benjamin D Singer
- Northwestern University Feinberg School of Medicine, Medicine, Chicago, IL, United States
| | - Franco D'Alessio
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, United States
| | - Alan Scott
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
308
|
Genes directly regulated by NF-κB in human hepatocellular carcinoma HepG2. Int J Biochem Cell Biol 2017; 89:157-170. [PMID: 28579529 DOI: 10.1016/j.biocel.2017.05.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/25/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
It has been well-known that over activation of NF-κB has close relationship with hepatitis and hepatocellular carcinoma (HCC). However, the complete and exact underlying molecular pathways and mechanisms still remain not fully understood. By manipulating NF-κB activity with its recognized activator TNFα and using ChIP-seq and RNA-seq techniques, this study identified 699 NF-κB direct target genes (DTGs) in a widely used HCC cell line, HepG2, including 399 activated and 300 repressed genes. In these NF-κB DTGs, 216 genes (126 activated and 90 repressed genes) are among the current HCC gene signature. In comparison with NF-κB target genes identified in LPS-induced THP-1 and TNFα-induced HeLa cells, only limited numbers (24-46) of genes were shared by the two cell lines, indicating the HCC specificity of identified genes. Functional annotation revealed that NF-κB DTGs in HepG2 cell are mainly related with many typical NF-κB-related biological processes including immune system process, response to stress, response to stimulus, defense response, and cell death, and signaling pathways of MAPK, TNF, TGF-beta, Chemokine, NF-kappa B, and Toll-like receptor. Some NF-κB DTGs are also involved in Hepatitis C and B pathways. It was found that 82 NF-κB DTGs code secretory proteins, which include CCL2 and DKK1 that have already been used as HCC markers. Finally, the NF-κB DTGs were further confirmed by detecting the NF-κB binding and expression of 14 genes with ChIP-PCR and RT-PCR. This study thus provides a useful NF-κB DTG list for future studies of NF-κB-related molecular mechanisms and theranostic biomarkers of HCC.
Collapse
|
309
|
Tariq M, Zhang J, Liang G, Ding L, He Q, Yang B. Macrophage Polarization: Anti-Cancer Strategies to Target Tumor-Associated Macrophage in Breast Cancer. J Cell Biochem 2017; 118:2484-2501. [DOI: 10.1002/jcb.25895] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 01/18/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Muhammad Tariq
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; Institute of Pharmacology and Toxicology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| | - Jieqiong Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; Institute of Pharmacology and Toxicology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| | - Guikai Liang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; Institute of Pharmacology and Toxicology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; Institute of Pharmacology and Toxicology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| | - Qiaojun He
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; Institute of Pharmacology and Toxicology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research; Institute of Pharmacology and Toxicology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou 310058 China
| |
Collapse
|
310
|
Cai H, Zhu XD, Ao JY, Ye BG, Zhang YY, Chai ZT, Wang CH, Shi WK, Cao MQ, Li XL, Sun HC. Colony-stimulating factor-1-induced AIF1 expression in tumor-associated macrophages enhances the progression of hepatocellular carcinoma. Oncoimmunology 2017; 6:e1333213. [PMID: 28932635 DOI: 10.1080/2162402x.2017.1333213] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/16/2017] [Accepted: 05/17/2017] [Indexed: 02/07/2023] Open
Abstract
M2-polarized (alternatively activated) macrophages play an important role in the progression of hepatocellular carcinoma (HCC). Allograft inflammatory factor 1 (AIF1) is overexpressed in M2-polarized macrophages. This study explored the role of AIF1 in tumor-associated macrophages in HCC. Macrophages were stimulated with colony-stimulating factor 1 (CSF1) to characterize the regulatory pathway of AIF1 in macrophages. The chromatin immunoprecipitation and luciferase reporter gene assay were conducted to examine transcription factors associated with AIF1 expression. AIF1 was down or upregulated, and the effects on tumor progression were evaluated by using in vitro and in vivo co-culture systems. A cytokine array was performed to screen the downstream functional components of AIF1. Tumor tissue from 206 patients with HCC were used to explore the clinical significance of AIF1. AIF1 induced a M2-like phenotype of macrophages. By facilitating the binding of c-Jun to the promoter of AIF1, CSF1 secreted from hepatoma cells increased AIF1 expression through the CSF1R-MEK1/2-Erk1/2-c-Jun axis. AIF1 expressed in macrophages promoted the migration of hepatoma cells in co-culture system of RAW264.7 and Hepa1-6 and tumor growth in an animal model. The cytokine array showed that CXCL16 was increased in RAW264.7 cells with overexpressed AIF1, leading to enhanced tumor cell migration. In human HCC tissue, AIF1-positive macrophages in the adjacent microenvironment was associated with microvascular invasion and advanced TNM stages and with patients' overall and disease-free survival (p = 0.002 for both). AIF1 expression in macrophages plays a pivotal role in the interaction between macrophages and hepatoma cells.
Collapse
Affiliation(s)
- Hao Cai
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Jian-Yang Ao
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bo-Gen Ye
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China.,Department of Organ Transplantation, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yuan-Yuan Zhang
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Zong-Tao Chai
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Cheng-Hao Wang
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Wen-Kai Shi
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Man-Qing Cao
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xiao-Long Li
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| |
Collapse
|
311
|
Abstract
BACKGROUND Early prognostic markers that identify high-risk patients could lead to increased surveillance, personalized immunosuppression, and improved long-term outcomes. The goal of this study was to validate 6-month urinary chemokine ligand 2 (CCL2) as a noninvasive predictor of long-term outcomes and compare it with 6-month urinary CXCL10. METHODS A prospective, observational renal transplant cohort (n = 185; minimum, 5-year follow-up) was evaluated. The primary composite outcome included 1 or more: allograft loss, renal function decline (>20% decrease estimated glomerular filtration rate between 6 months and last follow-up), and biopsy-proven rejection after 6 months. CCL2/CXCL10 are reported in relation to urine creatinine (ng/mmol). RESULTS Fifty-two patients (52/185, 28%) reached the primary outcome at a median 6.0 years, and their urinary CCL2:Cr was significantly higher compared with patients with stable allograft function (median [interquartile range], 38.6 ng/mmol [19.7-72.5] vs 25.9 ng/mmol [16.1-45.8], P = 0.009). Low urinary CCL2:Cr (≤70.0 ng/mmol) was associated with 88% 5-year event-free survival compared with 50% with high urinary CCL2:Cr (P < 0.0001). In a multivariate Cox-regression model, the only independent predictors of the primary outcome were high CCL2:Cr (hazard ratio [HR], 2.86; 95% confidence interval [95% CI], 1.33-5.73) and CXCL10:Cr (HR, 2.35; 95% CI, 1.23-4.88; both P = 0.009). Urinary CCL2:Cr/CXCL10:Cr area under the curves were 0.62 (P = 0.001)/0.63 (P = 0.03), respectively. Time-to-endpoint analysis according to combined high or low urinary chemokines demonstrates that endpoint-free survival depends on the overall early chemokine burden. CONCLUSIONS This study confirms that urinary CCL2:Cr is an independent predictor of long-term allograft outcomes. Urinary CCL2:Cr/CXCL10:Cr alone have similar prognostic performance, but when both are elevated, this suggests a worse prognosis. Therefore, urinary chemokines may be a useful tool for timely identification of high-risk patients.
Collapse
|
312
|
Gómez-Aristizábal A, Sharma A, Bakooshli MA, Kapoor M, Gilbert PM, Viswanathan S, Gandhi R. Stage-specific differences in secretory profile of mesenchymal stromal cells (MSCs) subjected to early- vs late-stage OA synovial fluid. Osteoarthritis Cartilage 2017; 25:737-741. [PMID: 27894935 DOI: 10.1016/j.joca.2016.11.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/17/2016] [Accepted: 11/21/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Although, mesenchymal stromal cells (MSCs) are being clinically investigated for their use in osteoarthritis (OA), it is unclear whether their postulated therapeutic properties are equally effective in the early- and late-stages of OA. In this study we investigated MSC cytokine secretion post-exposure to synovial fluid (SF), obtained from early- vs late-stage knee OA patients to justify a potential patient stratification strategy to maximize MSC-mediated treatment effects. METHOD Subjects were recruited and categorized into early- [Kellgren-Lawrence (KL) grade I/II, n = 12] and late-stage (KL-III/IV, n = 12) knee OA groups. SF samples were obtained, and their proteome was tested using multiplex assays, after 3-days culture, with and without MSCs. SFs cultured without MSCs were used as a baseline to identify MSC-secreted factors into SFs cultured with MSCs. Linear mixed-effect models and non-parametric tests were used to identify alterations in the MSC secretome during exposure to OA SF (3-days). MSCs cultured for 3-days in 0.5% fetal bovine serum (FBS)-supplemented medium were used to compare SF results with culture medium. RESULTS Following exposure to OA SF, the MSC secretome contained proteins that are involved in tissue repair, angiogenesis, chemotaxis, matrix remodeling and the clotting process. However, chemokine (C-X-C motif) ligand-8 (CXCL8; chemoattractant), interleukin-6 (IL6) and chemokine (C-C motif) ligand 2 (CCL2) were elevated in the MSC-secretome in response to early- vs late-stage OA SF. CONCLUSION Early- vs late-stage OA SF samples elicit a differential MSC secretome response, arguing for stratification of OA patients to maximize MSC-mediated therapeutic effects.
Collapse
Affiliation(s)
- A Gómez-Aristizábal
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada; The Arthritis Program, Toronto Western Hospital, Toronto, ON, Canada
| | - A Sharma
- The Arthritis Program, Toronto Western Hospital, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada
| | - M A Bakooshli
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, ON, Canada
| | - M Kapoor
- The Arthritis Program, Toronto Western Hospital, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada
| | - P M Gilbert
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada; Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, ON, Canada; Department of Biochemistry, University of Toronto, ON, Canada
| | - S Viswanathan
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada; The Arthritis Program, Toronto Western Hospital, Toronto, ON, Canada; Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada; Cell Therapy Program, University Health Network, Toronto, Canada.
| | - R Gandhi
- The Arthritis Program, Toronto Western Hospital, Toronto, ON, Canada; Division of Orthopaedic Surgery, Toronto Western Hospital, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
313
|
Valatas V, Filidou E, Drygiannakis I, Kolios G. Stromal and immune cells in gut fibrosis: the myofibroblast and the scarface. Ann Gastroenterol 2017; 30:393-404. [PMID: 28655975 PMCID: PMC5479991 DOI: 10.20524/aog.2017.0146] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/19/2017] [Indexed: 02/07/2023] Open
Abstract
Post-inflammatory scarring is the end-result of excessive extracellular matrix (ECM) accumulation and tissue architectural destruction. It represents a failure to effectively remodel ECM and achieve proper reinstitution and healing during chronic relapsing inflammatory processes. Scarring may affect the functionality of any organ, and in the case of inflammatory bowel disease (IBD)-associated fibrosis leads to stricture formation and often surgery to remove the affected bowel. The activated myofibroblast is the final effector cell that overproduces ECM under the influence of various mediators generated by an intense interplay of classic and non-classic immune cells. This review focuses on how proinflammatory mediators from various sources produced in different stages of intestinal inflammation can form profibrotic pathways that eventually lead to tissue scarring through sustained activation of myofibroblasts.
Collapse
Affiliation(s)
- Vassilis Valatas
- Laboratory of Gastroenterology, Faculty of Medicine, University of Crete, Heraklion (Vassilis Valatas, Ioannis Drygiannakis)
| | - Eirini Filidou
- Laboratory of Pharmacology, School of Medicine, Democritus University of Thrace, Dragana, Alexandroupolis (Eirini Filidou, George Kolios), Greece
| | - Ioannis Drygiannakis
- Laboratory of Gastroenterology, Faculty of Medicine, University of Crete, Heraklion (Vassilis Valatas, Ioannis Drygiannakis)
| | - George Kolios
- Laboratory of Pharmacology, School of Medicine, Democritus University of Thrace, Dragana, Alexandroupolis (Eirini Filidou, George Kolios), Greece
| |
Collapse
|
314
|
Mourik BC, Lubberts E, de Steenwinkel JEM, Ottenhoff THM, Leenen PJM. Interactions between Type 1 Interferons and the Th17 Response in Tuberculosis: Lessons Learned from Autoimmune Diseases. Front Immunol 2017; 8:294. [PMID: 28424682 PMCID: PMC5380685 DOI: 10.3389/fimmu.2017.00294] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/01/2017] [Indexed: 01/04/2023] Open
Abstract
The classical paradigm of tuberculosis (TB) immunity, with a central protective role for Th1 responses and IFN-γ-stimulated cellular responses, has been challenged by unsatisfactory results of vaccine strategies aimed at enhancing Th1 immunity. Moreover, preclinical TB models have shown that increasing IFN-γ responses in the lungs is more damaging to the host than to the pathogen. Type 1 interferon signaling and altered Th17 responses have also been associated with active TB, but their functional roles in TB pathogenesis remain to be established. These two host responses have been studied in more detail in autoimmune diseases (AID) and show functional interactions that are of potential interest in TB immunity. In this review, we first identify the role of type 1 interferons and Th17 immunity in TB, followed by an overview of interactions between these responses observed in systemic AID. We discuss (i) the effects of GM-CSF-secreting Th17.1 cells and type 1 interferons on CCR2+ monocytes; (ii) convergence of IL-17 and type 1 interferon signaling on stimulating B-cell activating factor production and the central role of neutrophils in this process; and (iii) synergy between IL-17 and type 1 interferons in the generation and function of tertiary lymphoid structures and the associated follicular helper T-cell responses. Evaluation of these autoimmune-related pathways in TB pathogenesis provides a new perspective on recent developments in TB research.
Collapse
Affiliation(s)
- Bas C Mourik
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Erik Lubberts
- Department of Rheumatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jurriaan E M de Steenwinkel
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Tom H M Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - Pieter J M Leenen
- Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
315
|
Murakami S, Shahbazian D, Surana R, Zhang W, Chen H, Graham GT, White SM, Weiner LM, Yi C. Yes-associated protein mediates immune reprogramming in pancreatic ductal adenocarcinoma. Oncogene 2017; 36:1232-1244. [PMID: 27546622 PMCID: PMC5322249 DOI: 10.1038/onc.2016.288] [Citation(s) in RCA: 166] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 12/19/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by a high degree of inflammation and profound immune suppression. Here we identify Yes-associated protein (Yap) as a critical regulator of the immunosuppressive microenvironment in both mouse and human PDAC. Within Kras:p53 mutant pancreatic ductal cells, Yap drives the expression and secretion of multiple cytokines/chemokines, which in turn promote the differentiation and accumulation of myeloid-derived suppressor cells (MDSCs) both in vitro and in vivo. Pancreas-specific knockout of Yap or antibody-mediated depletion of MDSCs promoted macrophage reprogramming, reactivation of T cells, apoptosis of Kras mutant neoplastic ductal cells and pancreatic regeneration after acute pancreatitis. In primary human PDAC, YAP expression levels strongly correlate with an MDSC gene signature, and high expression of YAP or MDSC-related genes predicts decreased survival in PDAC patients. These results reveal multifaceted roles of YAP in PDAC pathogenesis and underscore its promise as a therapeutic target for this deadly disease.
Collapse
Affiliation(s)
- Shigekazu Murakami
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - David Shahbazian
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Rishi Surana
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Weiying Zhang
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hengye Chen
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Garrett T. Graham
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shannon M. White
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Louis M. Weiner
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Chunling Yi
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
316
|
Li P, Wang H, Shao Q, Kong B, Qu X. Fucoidan modulates cytokine production and migration of THP-1-derived macrophages via colony-stimulating factor-1. Mol Med Rep 2017; 15:2325-2332. [DOI: 10.3892/mmr.2017.6228] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 11/02/2016] [Indexed: 11/06/2022] Open
|
317
|
Hartwig T, Montinaro A, von Karstedt S, Sevko A, Surinova S, Chakravarthy A, Taraborrelli L, Draber P, Lafont E, Arce Vargas F, El-Bahrawy MA, Quezada SA, Walczak H. The TRAIL-Induced Cancer Secretome Promotes a Tumor-Supportive Immune Microenvironment via CCR2. Mol Cell 2017; 65:730-742.e5. [PMID: 28212753 PMCID: PMC5316415 DOI: 10.1016/j.molcel.2017.01.021] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/21/2016] [Accepted: 01/17/2017] [Indexed: 01/14/2023]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is known for specifically killing cancer cells, whereas in resistant cancers, TRAIL/TRAIL-R can promote metastasis via Rac1 and PI3K. It remains unknown, however, whether and to what extent TRAIL/TRAIL-R signaling in cancer cells can affect the immune microenvironment. Here we show that TRAIL-triggered cytokine secretion from TRAIL-resistant cancer cells is FADD dependent and identify the TRAIL-induced secretome to drive monocyte polarization to myeloid-derived suppressor cells (MDSCs) and M2-like macrophages. TRAIL-R suppression in tumor cells impaired CCL2 production and diminished both lung MDSC presence and tumor growth. In accordance, the receptor of CCL2, CCR2, is required to facilitate increased MDSC presence and tumor growth. Finally, TRAIL and CCL2 are co-regulated with MDSC/M2 markers in lung adenocarcinoma patients. Collectively, endogenous TRAIL/TRAIL-R-mediated CCL2 secretion promotes accumulation of tumor-supportive immune cells in the cancer microenvironment, thereby revealing a tumor-supportive immune-modulatory role of the TRAIL/TRAIL-R system in cancer biology.
Collapse
Affiliation(s)
- Torsten Hartwig
- Centre for Cell Death, Cancer, and Inflammation, Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Antonella Montinaro
- Centre for Cell Death, Cancer, and Inflammation, Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Silvia von Karstedt
- Centre for Cell Death, Cancer, and Inflammation, Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alexandra Sevko
- Centre for Cell Death, Cancer, and Inflammation, Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Silvia Surinova
- Centre for Cell Death, Cancer, and Inflammation, Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Ankur Chakravarthy
- Department of Oncology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Lucia Taraborrelli
- Centre for Cell Death, Cancer, and Inflammation, Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Peter Draber
- Centre for Cell Death, Cancer, and Inflammation, Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Elodie Lafont
- Centre for Cell Death, Cancer, and Inflammation, Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Frederick Arce Vargas
- Cancer Immunology Unit, Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Mona A El-Bahrawy
- Department of Histopathology, Imperial College London, London W12 0NN, UK
| | - Sergio A Quezada
- Cancer Immunology Unit, Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation, Department of Cancer Biology, UCL Cancer Institute, University College London, London WC1E 6DD, UK.
| |
Collapse
|
318
|
Severino P, Palomino DT, Alvarenga H, Almeida CB, Pasqualim DC, Cury A, Salvalaggio PR, De Vasconcelos Macedo AL, Andrade MC, Aloia T, Bromberg S, Rizzo LV, Rocha FA, Marti LC. Human Lymph Node-Derived Fibroblastic and Double-Negative Reticular Cells Alter Their Chemokines and Cytokines Expression Profile Following Inflammatory Stimuli. Front Immunol 2017; 8:141. [PMID: 28261205 PMCID: PMC5307266 DOI: 10.3389/fimmu.2017.00141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 01/27/2017] [Indexed: 12/17/2022] Open
Abstract
Lymph node (LN) is a secondary lymphoid organ with highly organized and compartmentalized structure. LNs harbor B, T, and other cells among fibroblastic reticular cells (FRCs). FRCs are characterized by both podoplanin (PDPN/gp38) expression and by the lack of CD31 expression. FRCs are involved in several immune response processes but mechanisms underlying their function are still under investigation. Double-negative cells (DNCs), another cell population within LNs, are even less understood. They do not express PDPN or CD31, their localization within the LN is unknown, and their phenotype and function remain to be elucidated. This study evaluates the gene expression and cytokines and chemokines profile of human LN-derived FRCs and DNCs during homeostasis and following inflammatory stimuli. Cytokines and chemokines secreted by human FRCs and DNCs partially diverged from those identified in murine models that used similar stimulation. Cytokine and chemokine secretion and their receptors expression levels differed between stimulated DNCs and FRCs, with FRCs expressing a broader range of chemokines. Additionally, dendritic cells demonstrated increased migration toward FRCs, possibly due to chemokine-induced chemotaxis since migration was significantly decreased upon neutralization of secreted CCL2 and CCL20. Our study contributes to the understanding of the biology and functions of FRCs and DNCs and, accordingly, of the mechanisms involving them in immune cells activation and migration.
Collapse
Affiliation(s)
- Patricia Severino
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein , São Paulo , Brazil
| | - Diana Torres Palomino
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil; Programa de Alergia e Imunopatologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Heliene Alvarenga
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil; Programa de Alergia e Imunopatologia, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Camila Bononi Almeida
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein , São Paulo , Brazil
| | | | - Adriano Cury
- Hospital Israelita Albert Einstein, São Paulo, Brazil; Endocrinology Department, Santa Casa de Misericórdia de Sao Paulo, São Paulo, Brazil
| | - Paolo Rogério Salvalaggio
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein, São Paulo, Brazil; Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Maria Claudina Andrade
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein , São Paulo , Brazil
| | - Thiago Aloia
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein , São Paulo , Brazil
| | | | - Luiz Vicente Rizzo
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein , São Paulo , Brazil
| | - Fernanda Agostini Rocha
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein , São Paulo , Brazil
| | - Luciana C Marti
- Albert Einstein Research and Education Institute, Hospital Israelita Albert Einstein , São Paulo , Brazil
| |
Collapse
|
319
|
Liao CC, Day YJ, Lee HC, Liou JT, Chou AH, Liu FC. ERK Signaling Pathway Plays a Key Role in Baicalin Protection Against Acetaminophen-Induced Liver Injury. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:105-121. [DOI: 10.1142/s0192415x17500082] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Acetaminophen (APAP) overdose causes hepatocytes necrosis and acute liver failure. Baicalin (BA), a major flavonoid of Scutellariae radix, has potent hepatoprotective properties in traditional medicine. In the present study, we investigated the protective effects of BA on a APAP-induced liver injury in a mouse model. The mice received an intraperitoneal hepatotoxic dose of APAP (300[Formula: see text]mg/kg) and after 30[Formula: see text]min, were treated with BA at concentrations of 0, 15, 30, or 60[Formula: see text]mg/kg. After 16[Formula: see text]h of treatment, the mice were sacrificed for further analysis. APAP administration significantly elevated the serum alanine transferase (ALT) enzyme levels and hepatic myeloperoxidase (MPO) activity when compared with control animals. Baicalin treatment significantly attenuated the elevation of liver ALT levels, as well as hepatic MPO activity in a dose- dependent manner (15–60[Formula: see text]mg/kg) in APAP-treated mice. The strongest beneficial effects of BA were seen at a dose of 30[Formula: see text]mg/kg. BA treatment at 30[Formula: see text]mg/kg after APAP overdose reduced elevated hepatic cytokine (TNF-[Formula: see text] and IL-6) levels, and macrophage recruitment around the area of hepatotoxicity in immunohistochemical staining. Significantly, BA treatment can also decrease hepatic phosphorylated extracellular signal-regulated kinase (ERK) expression, which is induced by APAP overdose. Our data suggests that baicalin treatment can effectively attenuate APAP-induced liver injury by down-regulating the ERK signaling pathway and its downstream effectors of inflammatory responses. These results support that baicalin is a potential hepatoprotective agent.
Collapse
Affiliation(s)
- Chia-Chih Liao
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Yuan-Ji Day
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chen Lee
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Jiin-Tarng Liou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - An-Hsun Chou
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Fu-Chao Liu
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
320
|
Cuevas VD, Anta L, Samaniego R, Orta-Zavalza E, Vladimir de la Rosa J, Baujat G, Domínguez-Soto Á, Sánchez-Mateos P, Escribese MM, Castrillo A, Cormier-Daire V, Vega MA, Corbí ÁL. MAFB Determines Human Macrophage Anti-Inflammatory Polarization: Relevance for the Pathogenic Mechanisms Operating in Multicentric Carpotarsal Osteolysis. THE JOURNAL OF IMMUNOLOGY 2017; 198:2070-2081. [PMID: 28093525 DOI: 10.4049/jimmunol.1601667] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/16/2016] [Indexed: 12/13/2022]
Abstract
Macrophage phenotypic and functional heterogeneity derives from tissue-specific transcriptional signatures shaped by the local microenvironment. Most studies addressing the molecular basis for macrophage heterogeneity have focused on murine cells, whereas the factors controlling the functional specialization of human macrophages are less known. M-CSF drives the generation of human monocyte-derived macrophages with a potent anti-inflammatory activity upon stimulation. We now report that knockdown of MAFB impairs the acquisition of the anti-inflammatory profile of human macrophages, identify the MAFB-dependent gene signature in human macrophages and illustrate the coexpression of MAFB and MAFB-target genes in CD163+ tissue-resident and tumor-associated macrophages. The contribution of MAFB to the homeostatic/anti-inflammatory macrophage profile is further supported by the skewed polarization of monocyte-derived macrophages from multicentric carpotarsal osteolysis (Online Mendelian Inheritance in Man #166300), a pathology caused by mutations in the MAFB gene. Our results demonstrate that MAFB critically determines the acquisition of the anti-inflammatory transcriptional and functional profiles of human macrophages.
Collapse
Affiliation(s)
- Víctor D Cuevas
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain
| | - Laura Anta
- Servicio de Cirugía Ortopédica y Traumatología, Complejo Hospitalario de Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Rafael Samaniego
- Laboratorio de Inmuno-Oncología, Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
| | - Emmanuel Orta-Zavalza
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain
| | - Juan Vladimir de la Rosa
- Instituto de Investigaciones Biomedicas Alberto Sols, Consejo Superior de Investigaciones Científicas, 28029 Madrid, Spain
| | - Geneviève Baujat
- Unidad de Biomedicina, Instituto de Investigaciones Biomédicas-Universidad de Las Palmas de Gran Canaria (ULPGC), Instituto Universitario de Investigaciones Biomedicas y Sanitarias de la ULPGC, 35001 Las Palmas, Spain.,Département de Génétique, INSERM U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker Enfants Malades, 75015 Paris, France; and
| | - Ángeles Domínguez-Soto
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain
| | - Paloma Sánchez-Mateos
- Laboratorio de Inmuno-Oncología, Unidad de Microscopía Confocal, Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
| | - María M Escribese
- Institute for Applied Molecular Medicine, School of Medicine, University CEU San Pablo, Madrid, Spain
| | - Antonio Castrillo
- Instituto de Investigaciones Biomedicas Alberto Sols, Consejo Superior de Investigaciones Científicas, 28029 Madrid, Spain
| | - Valérie Cormier-Daire
- Unidad de Biomedicina, Instituto de Investigaciones Biomédicas-Universidad de Las Palmas de Gran Canaria (ULPGC), Instituto Universitario de Investigaciones Biomedicas y Sanitarias de la ULPGC, 35001 Las Palmas, Spain.,Département de Génétique, INSERM U781, Université Paris Descartes-Sorbonne Paris Cité, Institut Imagine, Hôpital Necker Enfants Malades, 75015 Paris, France; and
| | - Miguel A Vega
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain;
| | - Ángel L Corbí
- Laboratorio de Células Mieloides, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040 Madrid, Spain;
| |
Collapse
|
321
|
Kim YH, Lee SH. Mitochondrial reactive oxygen species regulate fungal protease-induced inflammatory responses. Toxicology 2017; 378:86-94. [PMID: 28087464 DOI: 10.1016/j.tox.2017.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 01/27/2023]
Abstract
Epidemiological studies have shown that fungal infections are a main cause of respiratory tract diseases, such as asthma, bronchopneumonia, intoxication, and invasive fungal disease. Fungi such as Aspergillus and Candida species have become increasingly important pathogens as the global climate changes. Accordingly, in this study, we evaluated the toxicological potential of Aspergillus protease in the lower respiratory tract. Exposure of Aspergillus protease to A549 cells induced upregulation of tumor necrosis factor (TNF)-α, monocyte chemoattractant protein (MCP)-1, and intercellular adhesion molecule (ICAM)-1 mRNAs and increased production of interleukin (IL)-8 and MCP-1 protein through enhanced mitochondrial reactive oxygen species (ROS) generation and activation of mitogen-activated protein kinase (MAPK) and activator protein (AP)-1. Furthermore, the mitochondrial ROS scavenger Mito-TEMPO, which inhibited MAPK and AP-1, significantly reduced MCP-1 and IL-1β mRNA expression and reduced HL-60 cell migration through the suppression of MCP-1 and IL-8 protein secretion. Thus, our results demonstrated that mitochondria were an important source of Aspergillus protease-stimulated ROS and that regulation of mitochondrial ROS modulated inflammatory responses by preventing activation of MAPK and AP-1 in A549 cells.
Collapse
Affiliation(s)
- Yun Hee Kim
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea; Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), Daejeon 34054, Republic of Korea
| | - Seung-Hyo Lee
- Graduate School of Medical Science and Engineering, Biomedical Research Center, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea.
| |
Collapse
|
322
|
Li X, Yao W, Yuan Y, Chen P, Li B, Li J, Chu R, Song H, Xie D, Jiang X, Wang H. Targeting of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy against hepatocellular carcinoma. Gut 2017; 66:157-167. [PMID: 26452628 DOI: 10.1136/gutjnl-2015-310514] [Citation(s) in RCA: 519] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 09/02/2015] [Accepted: 09/14/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is an aggressive malignancy with limited effective treatment options. An alternative strategy is to target cells, such as tumour-infiltrating macrophages, in the HCC tumour microenvironment. The CCL2/CCR2 axis is required for recruitment of monocytes/macrophages and is implicated in various aspects of liver pathology, including HCC. We investigated the feasibility of CCL2/CCR2 as a therapeutic target against HCC. DESIGN CCL2 expression was analysed in two independent HCC cohorts. Growth of three murine HCC cells was evaluated in an orthotopic model, a postsurgical recurrence model and a subcutaneous model in mice after blocking CCL2/CCR2 axis by a novel CCR2 antagonist or knocking out of host CCR2. In vivo macrophage or T cell depletion and in vitro cell coculture were further conducted to investigate CCL2/CCR2-mediated crosstalk between tumour-associated macrophages (TAMs) and tumour cells. RESULT CCL2 is overexpressed in human liver cancers and is prognostic for patients with HCC. Blockade of CCL2/CCR2 signalling with knockout of CCR2 or with a CCR2 antagonist inhibits malignant growth and metastasis, reduces postsurgical recurrence, and enhances survival. Further, therapeutic blocking of the CCL2/CCR2 axis inhibits the recruitment of inflammatory monocytes, infiltration and M2-polarisation of TAMs, resulting in reversal of the immunosuppression status of the tumour microenvironment and activation of an antitumorous CD8+ T cell response. CONCLUSIONS In patients with liver cancer, CCL2 is highly expressed and is a prognostic factor. Blockade of CCL2/CCR2 signalling suppresses murine liver tumour growth via activating T cell antitumour immune response. The results demonstrate the translational potential of CCL2/CCR2 blockade for treatment of HCCs.
Collapse
Affiliation(s)
- Xiaoguang Li
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenbo Yao
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ya Yuan
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Peizhan Chen
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bin Li
- The Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jingquan Li
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Ruiai Chu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haiyun Song
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China
| | - Dong Xie
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Xiaoqing Jiang
- The Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Hui Wang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.,Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China.,School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| |
Collapse
|
323
|
Puré E, Lo A. Can Targeting Stroma Pave the Way to Enhanced Antitumor Immunity and Immunotherapy of Solid Tumors? Cancer Immunol Res 2016; 4:269-78. [PMID: 27036971 DOI: 10.1158/2326-6066.cir-16-0011] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Solid tumors are complex organ-like structures. The potential of normal neighboring cells to contribute to the initiation, progression, and metastasis of epithelial-derived carcinomas has long been appreciated. However, the role of host cells has proven complex. Through multiple local and systemic mechanisms, nontransformed host cells can promote transition from a tumor-resistant to tumor-permissive environment, drive neoplastic transformation of epithelial cells, promote tumor growth, progression, and metastasis, but also constrain tumorigenesis. This complexity reflects the spatially and temporally dynamic involvement of multiple cell types and processes, including the development and recruitment of inflammatory, immune, endothelial, and mesenchymal stromal cells, and the remodeling of extracellular matrix. Our mechanistic understanding, as well as our ability to translate advances in our understanding of these mechanisms for therapeutic benefit, is rapidly advancing. Further insights will depend on delineating pathways that mediate the communication networks between inflammatory and immune cells with tumor and mesenchymal stromal cells and extracellular matrix. Here, we discuss the diversity of mesenchymal stromal cell populations and how context can dictate either their promotion or constraint of tumorigenesis. We review evidence for plasticity that allows for reprogramming of stromal cells and how tumor immunogenicity and desmoplasia influence the balance of immune-independent and immune-dependent regulation of tumor growth. The pivotal roles of matrix and mesenchymal stromal cells in modulating inflammation, antitumor immunity, and the efficacy of immune-based therapies are discussed. These concepts have emerged from data obtained from tumors of multiple organs, but we focus mostly on studies of pancreatic ductal adenocarcinomas.
Collapse
Affiliation(s)
- Ellen Puré
- University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Albert Lo
- University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
324
|
MCP1 triggers monocyte dysfunctions during abnormal osteogenic differentiation of mesenchymal stem cells in ankylosing spondylitis. J Mol Med (Berl) 2016; 95:143-154. [PMID: 27921117 DOI: 10.1007/s00109-016-1489-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/24/2016] [Accepted: 11/07/2016] [Indexed: 12/22/2022]
Abstract
Ankylosing spondylitis (AS) is a chronic inflammatory disease characterized by pathological osteogenesis and inflammation. However, the pathogenesis of AS and the pathological relationship between osteogenesis and inflammation in this disease remain largely unknown. Mesenchymal stem cells (MSCs) are multipotent progenitor cells capable of osteogenic differentiation and immunoregulation. Recently, we demonstrated that MSCs from AS patients (ASMSCs) have a greater potential for osteogenic differentiation than MSCs from healthy donors (HDMSCs), which therefore seems to be a component of pathological osteogenesis in AS. Previous studies have indicated that the immunoregulatory abilities of MSCs change following differentiation. However, the subsequent effects of ASMSCs during abnormal osteogenic differentiation are unclear. Here, we further demonstrated that ASMSCs secreted more monocyte chemoattractant protein 1 (MCP1) than HDMSCs during osteogenic differentiation. This enhanced MCP1 secretion augmented monocyte migration, increased classical macrophage polarization, and enhanced TNF-α secretion. Inhibiting MCP1 secretion from osteogenic differentiated ASMSCs using lentiviruses encoding short hairpin RNAs ameliorated these dysfunctions. Blocking the ERK1/2 pathway in ASMSCs with U0126 corrected the abnormal osteogenic differentiation, inhibited MCP1 overexpression, and prevented subsequent monocyte dysfunction. Finally, MCP1 expression was up-regulated during osteogenic differentiation in ASMSCs in vivo and was locally augmented in osteoblasts at ossification sites in AS patients. In summary, our study determined that MCP1 overexpression during abnormal osteogenic differentiation of ASMSCs triggers monocyte dysfunctions. We propose the novel hypothesis that pathological osteogenesis can lead to inflammation in AS. This hypothesis may contribute to reveal the precise pathological relationship between osteogenesis and inflammation in the field of osteoimmunology. KEY MESSAGE ASMSCs secreted more MCP1 during abnormal osteogenic differentiation. MCP1 overexpression leads to monocyte dysfunctions. Pathological osteogenesis can lead to inflammation in AS.
Collapse
|
325
|
Montenegro-Burke JR, Sutton JA, Rogers LM, Milne GL, McLean JA, Aronoff DM. Lipid profiling of polarized human monocyte-derived macrophages. Prostaglandins Other Lipid Mediat 2016; 127:1-8. [PMID: 27871801 PMCID: PMC6053630 DOI: 10.1016/j.prostaglandins.2016.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 11/11/2016] [Indexed: 12/31/2022]
Abstract
The highly orchestrated transcriptional and metabolic reprogramming during activation drastically transforms the main functions and physiology of human macrophages across the polarization spectrum. Lipids, for example, can modify protein function by acting remotely as signaling molecules but also locally by altering the physical properties of cellular membranes. These changes play key roles in the functions of highly plastic immune cells due to their involvement in inflammation, immune responses, phagocytosis and wound healing processes. We report an analysis of major membrane lipids of distinct phenotypes of resting (M0), classically activated (M1), alternatively activated (M2a) and deactivated (M2c) human monocyte derived macrophages from different donors. Samples were subjected to supercritical fluid chromatography-ion mobility-mass spectrometry analysis, which allowed separations based on lipid class, facilitating the profiling of their fatty acid composition. Different levels of arachidonic acid mobilization as well as other fatty acid changes were observed for different lipid classes in the distinct polarization phenotypes, suggesting the activation of highly orchestrated and specific enzymatic processes in the biosynthesis of lipid signaling molecules and cell membrane remodeling. Thromboxane A2 production appeared to be a specific marker of M1 polarization. These alterations to the global composition of lipid bi-layer membranes in the cell provide a potential methodology for the definition and determination of cellular and tissue activation states.
Collapse
Affiliation(s)
- J Rafael Montenegro-Burke
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37235, USA; Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, 37235, USA; Center for Innovative Technology, Vanderbilt University, Nashville, TN 37235, USA
| | - Jessica A Sutton
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Microbiology and Immunology, Meharry Medical College School of Medicine, Nashville, TN, 37208, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Lisa M Rogers
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ginger L Milne
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - John A McLean
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37235, USA; Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, 37235, USA; Center for Innovative Technology, Vanderbilt University, Nashville, TN 37235, USA
| | - David M Aronoff
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, TN, 37235, USA; Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA; Department of Microbiology and Immunology, Meharry Medical College School of Medicine, Nashville, TN, 37208, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
326
|
Ghosh AR, Bhattacharya R, Bhattacharya S, Nargis T, Rahaman O, Duttagupta P, Raychaudhuri D, Liu CSC, Roy S, Ghosh P, Khanna S, Chaudhuri T, Tantia O, Haak S, Bandyopadhyay S, Mukhopadhyay S, Chakrabarti P, Ganguly D. Adipose Recruitment and Activation of Plasmacytoid Dendritic Cells Fuel Metaflammation. Diabetes 2016; 65:3440-3452. [PMID: 27561727 DOI: 10.2337/db16-0331] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/19/2016] [Indexed: 11/13/2022]
Abstract
In obese individuals, visceral adipose tissue (VAT) is the seat of chronic low-grade inflammation (metaflammation), but the mechanistic link between increased adiposity and metaflammation largely remains unclear. In obese individuals, deregulation of a specific adipokine, chemerin, contributes to innate initiation of metaflammation by recruiting circulating plasmacytoid dendritic cells (pDCs) into VAT through chemokine-like receptor 1 (CMKLR1). Adipose tissue-derived high-mobility group B1 (HMGB1) protein activates Toll-like receptor 9 (TLR9) in the adipose-recruited pDCs by transporting extracellular DNA through receptor for advanced glycation end products (RAGE) and induces production of type I interferons (IFNs). Type I IFNs in turn help in proinflammatory polarization of adipose-resident macrophages. IFN signature gene expression in VAT correlates with both adipose tissue and systemic insulin resistance (IR) in obese individuals, which is represented by ADIPO-IR and HOMA2-IR, respectively, and defines two subgroups with different susceptibility to IR. Thus, this study reveals a pathway that drives adipose tissue inflammation and consequent IR in obesity.
Collapse
Affiliation(s)
- Amrit Raj Ghosh
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Roopkatha Bhattacharya
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Shamik Bhattacharya
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Titli Nargis
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Oindrila Rahaman
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Pritam Duttagupta
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Deblina Raychaudhuri
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Chinky Shiu Chen Liu
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Shounak Roy
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Parasar Ghosh
- Department of Rheumatology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | | | | | | | - Stefan Haak
- Zentrum Allergie und Umwelt, Technical University of Munich and Helmholtz Centre Munich, Munich, Germany
| | - Santu Bandyopadhyay
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Satinath Mukhopadhyay
- Department of Endocrinology, Institute of Postgraduate Medical Education and Research, Kolkata, India
| | - Partha Chakrabarti
- Division of Cell Biology and Physiology, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| | - Dipyaman Ganguly
- Division of Cancer Biology and Inflammatory Disorders, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
327
|
Ogiso H, Ito H, Ando T, Arioka Y, Kanbe A, Ando K, Ishikawa T, Saito K, Hara A, Moriwaki H, Shimizu M, Seishima M. The Deficiency of Indoleamine 2,3-Dioxygenase Aggravates the CCl4-Induced Liver Fibrosis in Mice. PLoS One 2016; 11:e0162183. [PMID: 27598994 PMCID: PMC5012673 DOI: 10.1371/journal.pone.0162183] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 08/18/2016] [Indexed: 12/11/2022] Open
Abstract
In the present study, we examined the role of indoleamine 2,3-dioxygenase (IDO) in the development of CCl4-induced hepatic fibrosis. The liver fibrosis induced by repetitive administration with CCl4 was aggravated in IDO-KO mice compared to WT mice. In IDO-KO mice treated with CCl4, the number of several inflammatory cells and the expression of pro-inflammatory cytokines increased in the liver. In the results, activated hepatic stellate cells (HSCs) and fibrogenic factors on HSCs increased after repetitive CCl4 administration in IDO-KO mice compared to WT mice. Moreover, the treatment with l-tryptophan aggravated the CCl4-induced hepatic fibrosis in WT mice. Our findings demonstrated that the IDO deficiency enhanced the inflammation in the liver and aggravated liver fibrosis in repetitive CCl4-treated mice.
Collapse
MESH Headings
- Alanine Transaminase/genetics
- Alanine Transaminase/immunology
- Animals
- Carbon Tetrachloride
- Cell Survival/drug effects
- Chemokine CCL2/genetics
- Chemokine CCL2/immunology
- Hepatic Stellate Cells/drug effects
- Hepatic Stellate Cells/immunology
- Hepatic Stellate Cells/pathology
- Hepatocytes/drug effects
- Hepatocytes/immunology
- Hepatocytes/pathology
- Immunophenotyping
- Indoleamine-Pyrrole 2,3,-Dioxygenase/deficiency
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Interleukin-1beta/genetics
- Interleukin-1beta/immunology
- Interleukin-6/genetics
- Interleukin-6/immunology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/pathology
- Liver/drug effects
- Liver/immunology
- Liver/pathology
- Liver Cirrhosis/chemically induced
- Liver Cirrhosis/genetics
- Liver Cirrhosis/immunology
- Liver Cirrhosis/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Proto-Oncogene Proteins c-sis/genetics
- Proto-Oncogene Proteins c-sis/immunology
- Tryptophan/pharmacology
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- Hideyuki Ogiso
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyasu Ito
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
- * E-mail:
| | - Tatsuya Ando
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yuko Arioka
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Ayumu Kanbe
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kazuki Ando
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tetsuya Ishikawa
- Department of Medical Technology, Nagoya University School of Health Sciences, Nagoya, Japan
| | - Kuniaki Saito
- Human Health Sciences, Kyoto University Graduate School of Medicine and Faculty of Medicine, Kyoto, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hisataka Moriwaki
- First Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masahito Shimizu
- First Department of Internal Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Mitsuru Seishima
- Department of Informative Clinical Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
328
|
Oliveira DCD, Hastreiter AA, Borelli P, Fock RA. The influence of protein malnutrition on the production of GM-CSF and M-CSF by macrophages. BRAZ J PHARM SCI 2016. [DOI: 10.1590/s1984-82502016000300003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
329
|
Li Y, Zheng Y, Li T, Wang Q, Qian J, Lu Y, Zhang M, Bi E, Yang M, Reu F, Yi Q, Cai Z. Chemokines CCL2, 3, 14 stimulate macrophage bone marrow homing, proliferation, and polarization in multiple myeloma. Oncotarget 2016; 6:24218-29. [PMID: 26155942 PMCID: PMC4695181 DOI: 10.18632/oncotarget.4523] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/05/2015] [Indexed: 12/14/2022] Open
Abstract
We previously showed that macrophages (MΦs) infiltrate the bone marrow (BM) of patients with myeloma and may play a role in drug resistance. This study analyzed chemokines expressed by myeloma BM that are responsible for recruiting monocytes to the tumor bed. We found that chemokines CCL3, CCL14, and CCL2 were highly expressed by myeloma and BM cells, and the levels of CCL14 and CCL3 in myeloma BM positively correlated with the percentage of BM-infiltrating MΦs. In vitro, these chemokines were responsible for chemoattracting human monocytes to tumor sites and in vivo for MΦ infiltration into myeloma-bearing BM in the 5TGM1 mouse model. Surprisingly, we also found that these chemokines stimulated MΦ in vitro proliferation induced by myeloma cells and in vivo in a human myeloma xenograft SCID mouse model. The chemokines also activated normal MΦ polarization and differentiation into myeloma-associated MΦs. Western blot analysis revealed that these chemokines promoted growth and survival signaling in MΦs via activating the PI3K/Akt and ERK MAPK pathways and c-myc expression. Thus, this study provides novel insight into the mechanism of MΦ infiltration of BM and also potential targets for improving the efficacy of chemotherapy in myeloma.
Collapse
Affiliation(s)
- Yi Li
- Bone Marrow Transplantation Center, Department of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yuhuan Zheng
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Hematology, Sichuan University, West China School of Medicine, Chengdu, Sichuan, China
| | - Tianshu Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Qiang Wang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jianfei Qian
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yong Lu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mingjun Zhang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Enguang Bi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Maojie Yang
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Frederic Reu
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Qing Yi
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zhen Cai
- Bone Marrow Transplantation Center, Department of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
330
|
Biswas SK, Bonecchi R. Colonic Macrophages "Remote Control" Adipose Tissue Inflammation and Insulin Resistance. Cell Metab 2016; 24:196-8. [PMID: 27508866 DOI: 10.1016/j.cmet.2016.07.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The early events linking diet-induced adipose tissue inflammation and insulin resistance remain poorly understood. In this issue of Cell Metabolism, Kawano et al. (2016) show that infiltration of colonic pro-inflammatory macrophages orchestrated by the intestinal CCL2/CCR2 axis kick-starts this process during high-fat-diet feeding.
Collapse
Affiliation(s)
- Subhra K Biswas
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A(∗)STAR), #04-06 Immunos, 8A Biomedical Grove, Singapore 138648, Singapore.
| | - Raffaella Bonecchi
- Humanitas Clinical and Research Center, Rozzano 20089, Italy; Department of Biomedical Sciences, Humanitas University, Rozzano 20089, Italy.
| |
Collapse
|
331
|
Kawano Y, Nakae J, Watanabe N, Kikuchi T, Tateya S, Tamori Y, Kaneko M, Abe T, Onodera M, Itoh H. Colonic Pro-inflammatory Macrophages Cause Insulin Resistance in an Intestinal Ccl2/Ccr2-Dependent Manner. Cell Metab 2016; 24:295-310. [PMID: 27508875 DOI: 10.1016/j.cmet.2016.07.009] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 03/22/2016] [Accepted: 07/15/2016] [Indexed: 12/27/2022]
Abstract
High-fat diet (HFD) induces low-grade chronic inflammation and insulin resistance. However, little is known about the mechanism underlying HFD-induced chronic inflammation in peripheral insulin-responsive tissues. Here, we show that colonic pro-inflammatory macrophages regulate insulin sensitivity under HFD conditions. To investigate the pathophysiological role of colonic macrophages, we generated macrophage-specific chemokine (C-C Motif) receptor 2 (Ccr2) knockout (M-Ccr2KO) and intestinal epithelial cell-specific tamoxifen-inducible Ccl2 knockout (Vil-Ccl2KO) mice. Both strains exhibited similar body weight to control under HFD. However, they exhibited decreased infiltration of colonic pro-inflammatory macrophages, decreased intestinal permeability, and inactivation of the colonic inflammasome. Interestingly, they showed significantly improved glucose tolerance and insulin sensitivity with decreased chronic inflammation of adipose tissue. Therefore, inhibition of pro-inflammatory macrophage infiltration prevents HFD-induced insulin resistance and could be a novel therapeutic approach for type 2 diabetes.
Collapse
Affiliation(s)
- Yoshinaga Kawano
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Jun Nakae
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan.
| | - Nobuyuki Watanabe
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-0074, Japan
| | - Tetsuhiro Kikuchi
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Sanshiro Tateya
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Yoshikazu Tamori
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; Department of Endocrinology and Metabolism, Chibune General Hospital, Osaka 555-0011, Japan
| | - Mari Kaneko
- Animal Resource Development Unit, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, Kobe 650-0047, Japan; Genetic Engineering Team, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, Kobe 650-0047, Japan
| | - Takaya Abe
- Genetic Engineering Team, Division of Bio-Function Dynamics Imaging, RIKEN Center for Life Science Technologies, Kobe 650-0047, Japan
| | - Masafumi Onodera
- Department of Human Genetics, National Center for Child Health and Development, Tokyo 157-0074, Japan
| | - Hiroshi Itoh
- Division of Endocrinology, Metabolism, and Nephrology, Department of Internal Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
332
|
Abstract
The human body combats infection and promotes wound healing through the remarkable process of inflammation. Inflammation is characterized by the recruitment of stromal cell activity including recruitment of immune cells and induction of angiogenesis. These cellular processes are regulated by a class of soluble molecules called cytokines. Based on function, cell target, and structure, cytokines are subdivided into several classes including: interleukins, chemokines, and lymphokines. While cytokines regulate normal physiological processes, chronic deregulation of cytokine expression and activity contributes to cancer in many ways. Gene polymorphisms of all types of cytokines are associated with risk of disease development. Deregulation RNA and protein expression of interleukins, chemokines, and lymphokines have been detected in many solid tumors and hematopoetic malignancies, correlating with poor patient prognosis. The current body of literature suggests that in some tumor types, interleukins and chemokines work against the human body by signaling to cancer cells and remodeling the local microenvironment to support the growth, survival, and invasion of primary tumors and enhance metastatic colonization. Some lymphokines are downregulated to suppress tumor progression by enhancing cytotoxic T cell activity and inhibiting tumor cell survival. In this review, we will describe the structure/function of several cytokine families and review our current understanding on the roles and mechanisms of cytokines in tumor progression. In addition, we will also discuss strategies for exploiting the expression and activity of cytokines in therapeutic intervention.
Collapse
Affiliation(s)
- M Yao
- University of Kansas Medical Center, Kansas City, KS, United States
| | - G Brummer
- University of Kansas Medical Center, Kansas City, KS, United States
| | - D Acevedo
- University of Kansas Medical Center, Kansas City, KS, United States
| | - N Cheng
- University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
333
|
Ding J, Chen B, Lv T, Liu X, Fu X, Wang Q, Yan L, Kang N, Cao Y, Xiao R. Bone Marrow Mesenchymal Stem Cell-Based Engineered Cartilage Ameliorates Polyglycolic Acid/Polylactic Acid Scaffold-Induced Inflammation Through M2 Polarization of Macrophages in a Pig Model. Stem Cells Transl Med 2016; 5:1079-89. [PMID: 27280797 DOI: 10.5966/sctm.2015-0263] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 03/14/2016] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED : The regeneration of tissue-engineered cartilage in an immunocompetent environment usually fails due to severe inflammation induced by the scaffold and their degradation products. In the present study, we compared the tissue remodeling and the inflammatory responses of engineered cartilage constructed with bone marrow mesenchymal stem cells (BMSCs), chondrocytes, or both and scaffold group in pigs. The cartilage-forming capacity of the constructs in vitro and in vivo was evaluated by histological, biochemical, and biomechanical analyses, and the inflammatory response was investigated by quantitative analysis of foreign body giant cells and macrophages. Our data revealed that BMSC-based engineered cartilage suppressed in vivo inflammation through the alteration of macrophage phenotype, resulting in better tissue survival compared with those regenerated with chondrocytes alone or in combination with BMSCs. To further confirm the macrophage phenotype, an in vitro coculture system established by engineered cartilage and macrophages was studied using immunofluorescence, enzyme-linked immunosorbent assay, and gene expression analysis. The results demonstrated that BMSC-based engineered cartilage promoted M2 polarization of macrophages with anti-inflammatory phenotypes including the upregulation of CD206, increased IL-10 synthesis, decreased IL-1β secretion, and alterations in gene expression indicative of M1 to M2 transition. It was suggested that BMSC-seeded constructs have the potential to ameliorate scaffold-induced inflammation and improve cartilaginous tissue regeneration through M2 polarization of macrophages. SIGNIFICANCE Finding a strategy that can prevent scaffold-induced inflammation is of utmost importance for the regeneration of tissue-engineered cartilage in an immunocompetent environment. This study demonstrated that bone marrow mesenchymal stem cell (BMSC)-based engineered cartilage could suppress inflammation by increasing M2 polarization of macrophages, resulting in better tissue survival in a pig model. Additionally, the effect of BMSC-based cartilage on the phenotype conversion of macrophages was further studied through an in vitro coculture system. This study could provide further support for the regeneration of cartilage engineering in immunocompetent animal models and provide new insight into the interaction of tissue-engineered cartilage and macrophages.
Collapse
Affiliation(s)
- Jinping Ding
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Bo Chen
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Tao Lv
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qian Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Li Yan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Ning Kang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yilin Cao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
334
|
Minutti CM, García-Fojeda B, Sáenz A, de las Casas-Engel M, Guillamat-Prats R, de Lorenzo A, Serrano-Mollar A, Corbí ÁL, Casals C. Surfactant Protein A Prevents IFN-γ/IFN-γ Receptor Interaction and Attenuates Classical Activation of Human Alveolar Macrophages. THE JOURNAL OF IMMUNOLOGY 2016; 197:590-8. [DOI: 10.4049/jimmunol.1501032] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 05/07/2016] [Indexed: 11/19/2022]
|
335
|
Iseri K, Iyoda M, Ohtaki H, Matsumoto K, Wada Y, Suzuki T, Yamamoto Y, Saito T, Hihara K, Tachibana S, Honda K, Shibata T. Therapeutic effects and mechanism of conditioned media from human mesenchymal stem cells on anti-GBM glomerulonephritis in WKY rats. Am J Physiol Renal Physiol 2016; 310:F1182-91. [DOI: 10.1152/ajprenal.00165.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/24/2016] [Indexed: 02/06/2023] Open
Abstract
Recent studies have demonstrated that conditioned media derived from mesenchymal stem cells (MSC-CM) have therapeutic effects in various experimental diseases. However, the therapeutic mechanism is not fully understood. In the present study, we investigated the therapeutic effects and mechanism of MSC-CM in experimental antiglomerular basement membrane glomerulonephritis. We administered either MSC-CM or vehicle from day 0 to day 10 after the induction of nephrotoxic serum nephritis in Wistar-Kyoto rats. In vitro, we analyzed the effects of MSC-CM on TNF-α-mediated cytokine production in cultured normal human mesangial cells, proximal tubular (HK-2) cells, human umbilical vein endothelial cells, and monocytes (THP-1 and peripheral blood mononuclear cells). Compared with vehicle treatment, MSC-CM treatment improved proteinuria and renal dysfunction. Histologically, MSC-CM-treated rats had reduced crescent formation and glomerular ED1+ macrophage infiltration and increased glomerular ED2+ macrophage infiltration. Increased serum monocyte chemoattractant protein (MCP)-1 levels were observed in MSC-CM-treated rats. Renal cortical mRNA expression levels of proinflammatory cytokines, such as TNF-α and IL-6, and of the T helper cell 1 cytokine interferon-γ were greatly decreased by MSC-CM treatment. In vitro, pretreatment with MSC-CM blocked TNF-α-mediated IL-8 release in normal human mesangial cells and HK-2 cells. TNF-α-mediated MCP-1 release was enhanced by pretreatment with MSC-CM in human umbilical vein endothelial cells and HK-2 cells and was strikingly enhanced in THP-1 cells. Stimulation of peripheral blood mononuclear cells with a combination of MCP-1 and IL-4 enhanced the expression of M2-associated genes compared with IL-4 alone. We demonstrated that MSC-CM had therapeutic effects in experimental antiglomerular basement membrane glomerulonephritis that were mediated through anti-inflammatory effects that were partly due to acceleration of M2 macrophage polarization, which might be mediated by MCP-1 enhancement.
Collapse
Affiliation(s)
- Ken Iseri
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Masayuki Iyoda
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Hirokazu Ohtaki
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Kei Matsumoto
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Yukihiro Wada
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Taihei Suzuki
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Yasutaka Yamamoto
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Tomohiro Saito
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Kei Hihara
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Shohei Tachibana
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| | - Kazuho Honda
- Department of Anatomy, Showa University School of Medicine, Tokyo, Japan
| | - Takanori Shibata
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan; and
| |
Collapse
|
336
|
Pont MJ, Honders MW, Kremer AN, van Kooten C, Out C, Hiemstra PS, de Boer HC, Jager MJ, Schmelzer E, Vries RG, Al Hinai AS, Kroes WG, Monajemi R, Goeman JJ, Böhringer S, Marijt WAF, Falkenburg JHF, Griffioen M. Microarray Gene Expression Analysis to Evaluate Cell Type Specific Expression of Targets Relevant for Immunotherapy of Hematological Malignancies. PLoS One 2016; 11:e0155165. [PMID: 27171398 PMCID: PMC4865094 DOI: 10.1371/journal.pone.0155165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 04/25/2016] [Indexed: 12/15/2022] Open
Abstract
Cellular immunotherapy has proven to be effective in the treatment of hematological cancers by donor lymphocyte infusion after allogeneic hematopoietic stem cell transplantation and more recently by targeted therapy with chimeric antigen or T-cell receptor-engineered T cells. However, dependent on the tissue distribution of the antigens that are targeted, anti-tumor responses can be accompanied by undesired side effects. Therefore, detailed tissue distribution analysis is essential to estimate potential efficacy and toxicity of candidate targets for immunotherapy of hematological malignancies. We performed microarray gene expression analysis of hematological malignancies of different origins, healthy hematopoietic cells and various non-hematopoietic cell types from organs that are often targeted in detrimental immune responses after allogeneic stem cell transplantation leading to graft-versus-host disease. Non-hematopoietic cells were also cultured in the presence of IFN-γ to analyze gene expression under inflammatory circumstances. Gene expression was investigated by Illumina HT12.0 microarrays and quality control analysis was performed to confirm the cell-type origin and exclude contamination of non-hematopoietic cell samples with peripheral blood cells. Microarray data were validated by quantitative RT-PCR showing strong correlations between both platforms. Detailed gene expression profiles were generated for various minor histocompatibility antigens and B-cell surface antigens to illustrate the value of the microarray dataset to estimate efficacy and toxicity of candidate targets for immunotherapy. In conclusion, our microarray database provides a relevant platform to analyze and select candidate antigens with hematopoietic (lineage)-restricted expression as potential targets for immunotherapy of hematological cancers.
Collapse
Affiliation(s)
- M. J. Pont
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - M. W. Honders
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - A. N. Kremer
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Internal Medicine 5, Hematology and Oncology, University Hospital Erlangen, Erlangen, Germany
| | - C. van Kooten
- Department of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - C. Out
- Department of Dermatology, Leiden University Medical Center, Leiden, the Netherlands
| | - P. S. Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | - H. C. de Boer
- Department of Nephrology and the Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - M. J. Jager
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - E. Schmelzer
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - R. G. Vries
- Hubrecht Institute for Developmental Biology and Stem Cell Research and University Medical Centre Utrecht, Utrecht, the Netherlands
| | - A. S. Al Hinai
- Department of Hematology, Erasmus University Medical Center Cancer Institute, Rotterdam, the Netherlands
| | - W. G. Kroes
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - R. Monajemi
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - J. J. Goeman
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
- Radboud Institute for Molecular Life Science, Radboud University Medical Center, Nijmegen, The Netherlands
| | - S. Böhringer
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - W. A. F. Marijt
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - J. H. F. Falkenburg
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - M. Griffioen
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
- * E-mail:
| |
Collapse
|
337
|
Paquinimod reduces skin fibrosis in tight skin 1 mice, an experimental model of systemic sclerosis. J Dermatol Sci 2016; 83:52-9. [PMID: 27156795 DOI: 10.1016/j.jdermsci.2016.04.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/08/2016] [Accepted: 04/18/2016] [Indexed: 11/21/2022]
Abstract
BACKGROUND Systemic Sclerosis (SSc) is an autoimmune disease characterized by vascular and immune dysfunction. A hallmark of SSc is the excessive accumulation of extracellular matrix in the skin and in internal organs. There is a high and unmet medical need for novel therapies in this disease. The pathogenesis of SSc is complex and still poorly understood, but the innate immune system has emerged as an important factor in the disease. SSc patients show increased numbers of macrophages/monocytes in the blood and in the skin compared to healthy individuals and these cells are important sources of profibrotic cytokines and chemokines. Paquinimod belongs to a class of orally active quinoline-3-carboxamide (quinoline) derivatives with immunomodulatory properties and has shown effects in several models of autoimmune/inflammatory disorders. Paquinimod is currently in clinical development for treatment of SSc. The immunomodulatory effects of paquinimod is by targeting the myeloid cell compartment via the S100A9 protein. OBJECTIVE In this study we investigate whether targeting of myeloid cells by paquinimod can effect disease development in an experimental model of SSc, the tight skin 1 (Tsk-1) mouse model. METHODS Seven weeks old female B6.Cg-Fbn1(Tsk)/J (Tsk-1) mice were treated with vehicle or paquinimod at the dose of 5 or 25mg/kg/day in the drinking water for 8 weeks. The effect of paquinimod on the level of skin fibrosis and on different subpopulations within the myeloid cell compartment in skin biopsies were evaluated by using histology, immunohistochemisty, a hydroxyproline assay and real-time PCR. Furthermore, the level of IgG in serum from treated animals was also analysed. The statistical analyses were performed using Mann-Whitney nonparametric two tailed rank test. RESULTS The results show that treatment with paquinimod reduces skin fibrosis measured as reduction of skin thickness and decreased number of myofibroblasts and total hydroxyproline content. The effect on fibrosis was associated with a polarization of macrophages in the skin from a pro-fibrotic M2 to a M1 phenotype. Paquinimod treatment also resulted in a reduced TGFβ-response in the skin and an abrogation of the increased auto-antibody production in this SSc model. CONCLUSIONS Paquinimod reduces skin fibrosis in an experimental model of SSc, and this effect correlates with local and systemic effects on the immune system.
Collapse
|
338
|
Palmer CS, Cherry CL, Sada-Ovalle I, Singh A, Crowe SM. Glucose Metabolism in T Cells and Monocytes: New Perspectives in HIV Pathogenesis. EBioMedicine 2016; 6:31-41. [PMID: 27211546 PMCID: PMC4856752 DOI: 10.1016/j.ebiom.2016.02.012] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 02/05/2016] [Accepted: 02/05/2016] [Indexed: 02/06/2023] Open
Abstract
Activation of the immune system occurs in response to the recognition of foreign antigens and receipt of optimal stimulatory signals by immune cells, a process that requires energy. Energy is also needed to support cellular growth, differentiation, proliferation, and effector functions of immune cells. In HIV-infected individuals, persistent viral replication, together with inflammatory stimuli contributes to chronic immune activation and oxidative stress. These conditions remain even in subjects with sustained virologic suppression on antiretroviral therapy. Here we highlight recent studies demonstrating the importance of metabolic pathways, particularly those involving glucose metabolism, in differentiation and maintenance of the activation states of T cells and monocytes. We also discuss how changes in the metabolic status of these cells may contribute to ongoing immune activation and inflammation in HIV- infected persons and how this may contribute to disease progression, establishment and persistence of the HIV reservoir, and the development of co-morbidities. We provide evidence that other viruses such as Epstein-Barr and Flu virus also disrupt the metabolic machinery of their host cells. Finally, we discuss how redox signaling mediated by oxidative stress may regulate metabolic responses in T cells and monocytes during HIV infection.
Collapse
Affiliation(s)
- Clovis S Palmer
- Centre for Biomedical Research, Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, Monash University, Melbourne, Australia.
| | - Catherine L Cherry
- Centre for Biomedical Research, Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, Monash University, Melbourne, Australia; Infectious Diseases Department, The Alfred Hospital, Melbourne, Australia; School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| | - Isabel Sada-Ovalle
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico
| | - Amit Singh
- Department of Microbiology and Cell Biology, Centre for Infectious Disease and Research (CIDR), Indian Institute of Science, India
| | - Suzanne M Crowe
- Centre for Biomedical Research, Burnet Institute, Melbourne, Australia; Department of Infectious Diseases, Monash University, Melbourne, Australia; Infectious Diseases Department, The Alfred Hospital, Melbourne, Australia
| |
Collapse
|
339
|
CCL2 Mediates Neuron-Macrophage Interactions to Drive Proregenerative Macrophage Activation Following Preconditioning Injury. J Neurosci 2016; 35:15934-47. [PMID: 26631474 DOI: 10.1523/jneurosci.1924-15.2015] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
CNS neurons in adult mammals do not spontaneously regenerate axons after spinal cord injury. Preconditioning peripheral nerve injury allows the dorsal root ganglia (DRG) sensory axons to regenerate beyond the injury site by promoting expression of regeneration-associated genes. We have previously shown that peripheral nerve injury increases the number of macrophages in the DRGs and that the activated macrophages are critical to the enhancement of intrinsic regeneration capacity. The present study identifies a novel chemokine signal mediated by CCL2 that links regenerating neurons with proregenerative macrophage activation. Neutralization of CCL2 abolished the neurite outgrowth activity of conditioned medium obtained from neuron-macrophage cocultures treated with cAMP. The neuron-macrophage interactions that produced outgrowth-promoting conditioned medium required CCL2 in neurons and CCR2/CCR4 in macrophages. The conditioning effects were abolished in CCL2-deficient mice at 3 and 7 d after sciatic nerve injury, but CCL2 was dispensable for the initial growth response and upregulation of GAP-43 at the 1 d time point. Intraganglionic injection of CCL2 mimicked conditioning injury by mobilizing M2-like macrophages. Finally, overexpression of CCL2 in DRGs promoted sensory axon regeneration in a rat spinal cord injury model without harmful side effects. Our data suggest that CCL2-mediated neuron-macrophage interaction plays a critical role for amplification and maintenance of enhanced regenerative capacity by preconditioning peripheral nerve injury. Manipulation of chemokine signaling mediating neuron-macrophage interactions may represent a novel therapeutic approach to promote axon regeneration after CNS injury.
Collapse
|
340
|
Singer BH, Newstead MW, Zeng X, Cooke CL, Thompson RC, Singer K, Ghantasala R, Parent JM, Murphy GG, Iwashyna TJ, Standiford TJ. Cecal Ligation and Puncture Results in Long-Term Central Nervous System Myeloid Inflammation. PLoS One 2016; 11:e0149136. [PMID: 26862765 PMCID: PMC4749127 DOI: 10.1371/journal.pone.0149136] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/26/2016] [Indexed: 12/24/2022] Open
Abstract
Survivors of sepsis often experience long-term cognitive and functional decline. Previous studies utilizing lipopolysaccharide injection and cecal ligation and puncture in rodent models of sepsis have demonstrated changes in depressive-like behavior and learning and memory after sepsis, as well as evidence of myeloid inflammation and cytokine expression in the brain, but the long-term course of neuroinflammation after sepsis remains unclear. Here, we utilize cecal ligation and puncture with greater than 80% survival as a model of sepsis. We found that sepsis survivor mice demonstrate deficits in extinction of conditioned fear, but no acquisition of fear conditioning, nearly two months after sepsis. These cognitive changes occur in the absence of neuronal loss or changes in synaptic density in the hippocampus. Sepsis also resulted in infiltration of monocytes and neutrophils into the CNS at least two weeks after sepsis in a CCR2 independent manner. Cellular inflammation is accompanied by long-term expression of pro-inflammatory cytokine and chemokine genes, including TNFα and CCR2 ligands, in whole brain homogenates. Gene expression analysis of microglia revealed that while microglia do express anti-microbial genes and damage-associated molecular pattern molecules of the S100A family of genes at least 2 weeks after sepsis, they do not express the cytokines observed in whole brain homogenates. Our results indicate that in a naturalistic model of infection, sepsis results in long-term neuroinflammation, and that this sustained inflammation is likely due to interactions among multiple cell types, including resident microglia and peripherally derived myeloid cells.
Collapse
Affiliation(s)
- Benjamin H. Singer
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail:
| | - Michael W. Newstead
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Xianying Zeng
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Christopher L. Cooke
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Robert C. Thompson
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Kanakadurga Singer
- Department of Pediatrics, Division of Endocrinology and Metabolism, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Ramya Ghantasala
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Jack M. Parent
- Department of Neurology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Geoffrey G. Murphy
- Molecular and Behavioral Neuroscience Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Theodore J. Iwashyna
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Center for Clinical Management Research, VA Ann Arbor Health System, Ann Arbor, Michigan, United States of America
| | - Theodore J. Standiford
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| |
Collapse
|
341
|
Wei L, Wang H, Yang F, Ding Q, Zhao J. Interleukin-17 potently increases non-small cell lung cancer growth. Mol Med Rep 2016; 13:1673-80. [PMID: 26708832 DOI: 10.3892/mmr.2015.4694] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 11/06/2015] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to explore the effects of interleukin (IL)-17 on the growth and metastasis of tumors that were subcutaneously implanted into C57BL/6 mice. Lewis lung carcinoma (LLC) cells were subcutaneously injected into C57BL/6 mice followed by intraperitoneal injection of mouse recombinant IL-17 protein (IL-17 groups) or phosphate-buffered saline (control groups). Tumor growth and metastasis were assessed by measuring the size and weight of tumors and cervical lymph nodes, respectively. Cytokine expression in tumor masses was quantified by reverse transcription-quantitative polymerase chain reaction and western blotting, respectively. CCR2-positive macrophage infiltration in tumor masses was detected by flow cytometric analysis. The proliferation and migration of LLC cells, stimulated by the IL-17 protein were detected by Cell Counting kit (CCK)-8 and wound scratch assays in vitro. Tumors were grafted into the C57BL/6 mice. The mice that were intraperitoneally injected with IL-17 exhibited significantly larger tumors compared with the control mice. After day 7 of injection and beyond, the weight of cervical lymph nodes in IL-17 groups was higher than that in the control mice. It was also demonstrated that the number of CCR2-positive macrophages that infiltrated the tumor masses in the IL-17 groups was higher than that of the control mice. CD34 expression in vascular endothelial cells was also higher in tumors grafted in IL-17 mice than those grafted in control mice. Furthermore, the tumor tissue mRNA and protein expression levels of vascular endothelial growth factor, matrix metalloproteinase (MMP)-2, MMP-9 and tumor necrosis factor-α were greater in mice from the IL-17 group than the control mice, while levels of migration inhibitory factor and thrombospondin-1 were lower in mice from the IL-17 group than in the control. IL-17 also increased the migration of LLC cells in vitro. In conclusion, IL-17 exhibited the ability to promote tumor growth by increasing angiogenesis, metastasis and increasing CCR2+ macrophage infiltration into tumors.
Collapse
MESH Headings
- Animals
- Antigens, CD34/metabolism
- Carcinoma, Non-Small-Cell Lung/blood supply
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Proliferation
- Gene Expression Regulation, Neoplastic
- Interleukin-17/metabolism
- Intramolecular Oxidoreductases/metabolism
- Lung Neoplasms/blood supply
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Macrophage Migration-Inhibitory Factors/metabolism
- Macrophages/pathology
- Mice, Inbred C57BL
- Neoplasm Metastasis
- Neoplasm Transplantation
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Receptors, CCR2/metabolism
- Thrombospondin 1/metabolism
Collapse
Affiliation(s)
- Lei Wei
- Department of Anesthesiology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Hui Wang
- Department of Anesthesiology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Fen Yang
- Department of Anesthesiology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Qi Ding
- Department of Anesthesiology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Jianhua Zhao
- Department of Anesthesiology, Suzhou Municipal Hospital Affiliated to Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| |
Collapse
|
342
|
Castillo EF, Ray AL, Beswick EJ. MK2: an unrecognized regulator of tumor promoting macrophages in colorectal cancer? ACTA ACUST UNITED AC 2016; 3. [PMID: 26998523 PMCID: PMC4798244 DOI: 10.14800/macrophage.1166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignancies and is associated closely with inflammation before and after development. Macrophages promote colitis and colitis-associated CRC. M1 macrophages contribute to colitis directly through the production of proinflammatory cytokines and through activation of proinflammatory immune cell phenotypes. In cancer, both M1 and M2 macrophages participate in tumor development and progression through cytokine production, changes in cell signaling and activation of T cells. We have identified the mitogen-activated protein kinase-activated protein kinase 2 (MK2) as a regulator of macrophages during colitis-associated CRC (CAC). MK2 is a proinflammatory kinase that promotes production of IL-1α, IL-1β, IL-6 and TNF-α. MK2−/− mice have decreases in macrophages, macrophage-associated chemokines, and proinflammatory cytokines. Most significantly, MK2−/− mice do not develop neoplasms in an inflammatory model of CRC. However, addition of MK2+/+ macrophages to MK2−/− mice increases production of proinflammatory cytokines. In wild type mice, both cytokines and tumor burdens increase upon addition of additional macrophages. These data support the importance of MK2 in macrophage regulation during inflammation-associated CRC.
Collapse
Affiliation(s)
- Eliseo F Castillo
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Anita L Ray
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Ellen J Beswick
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
343
|
González-Domínguez É, Domínguez-Soto Á, Nieto C, Flores-Sevilla JL, Pacheco-Blanco M, Campos-Peña V, Meraz-Ríos MA, Vega MA, Corbí ÁL, Sánchez-Torres C. Atypical Activin A and IL-10 Production Impairs Human CD16+ Monocyte Differentiation into Anti-Inflammatory Macrophages. THE JOURNAL OF IMMUNOLOGY 2016; 196:1327-37. [PMID: 26729812 DOI: 10.4049/jimmunol.1501177] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 11/23/2015] [Indexed: 12/24/2022]
Abstract
Human CD14(++)CD16(-) and CD14(+/lo)CD16(+) monocyte subsets comprise 85 and 15% of blood monocytes, respectively, and are thought to represent distinct stages in the monocyte differentiation pathway. However, the differentiation fates of both monocyte subsets along the macrophage (Mϕ) lineage have not yet been elucidated. We have now evaluated the potential of CD14(++) CD16(-) and CD16(+) monocytes to differentiate and to be primed toward pro- or anti-inflammatory Mϕs upon culture with GM-CSF or M-CSF, respectively (subsequently referred to as GM14, M14, GM16, or M16). Whereas GM16 and GM14 were phenotypic and functionally analogous, M16 displayed a more proinflammatory profile than did M14. Transcriptomic analyses evidenced that genes associated with M-CSF-driven Mϕ differentiation (including FOLR2, IL10, IGF1, and SERPINB2) are underrepresented in M16 with respect to M14. The preferential proinflammatory skewing of M16 relative to M14 was found to be mediated by the secretion of activin A and the low levels of IL-10 produced by M16. In fact, activin A receptor blockade during the M-CSF-driven differentiation of CD16(+) monocytes, or addition of IL-10-containing M14-conditioned medium, significantly enhanced their expression of anti-inflammatory-associated molecules while impairing their acquisition of proinflammatory-related markers. Thus, we propose that M-CSF drives CD14(++)CD16- monocyte differentiation into bona fide anti-inflammatory Mϕs in a self-autonomous manner, whereas M-CSF-treated CD16(+) monocytes generate Mϕs with a skewed proinflammatory profile by virtue of their high activin A expression unless additional anti-inflammatory stimuli such as IL-10 are provided.
Collapse
Affiliation(s)
- Érika González-Domínguez
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 07360 Mexico City, Mexico
| | - Ángeles Domínguez-Soto
- Laboratorio de Células Mieloides, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas, 28040 Madrid, Spain; and
| | - Concha Nieto
- Laboratorio de Células Mieloides, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas, 28040 Madrid, Spain; and
| | - José Luis Flores-Sevilla
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 07360 Mexico City, Mexico
| | - Mariana Pacheco-Blanco
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 07360 Mexico City, Mexico
| | - Victoria Campos-Peña
- Laboratorio Experimental de Enfermedades Neurodegenerativas, Instituto Nacional de Neurología y Neurocirugía "Manuel Velazco Suárez," 14269 Mexico City, Mexico
| | - Marco A Meraz-Ríos
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 07360 Mexico City, Mexico
| | - Miguel A Vega
- Laboratorio de Células Mieloides, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas, 28040 Madrid, Spain; and
| | - Ángel L Corbí
- Laboratorio de Células Mieloides, Departamento de Microbiología Molecular y Biología de las Infecciones, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Cientificas, 28040 Madrid, Spain; and
| | - Carmen Sánchez-Torres
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 07360 Mexico City, Mexico;
| |
Collapse
|
344
|
Kusmartsev S, Dominguez-Gutierrez PR, Canales BK, Bird VG, Vieweg J, Khan SR. Calcium Oxalate Stone Fragment and Crystal Phagocytosis by Human Macrophages. J Urol 2015; 195:1143-51. [PMID: 26626217 DOI: 10.1016/j.juro.2015.11.048] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2015] [Indexed: 01/28/2023]
Abstract
PURPOSE In murine and human hyperoxaluric conditions macrophages can be seen surrounding renal calcium oxalate crystal deposits. We hypothesized that macrophages have a role in degrading and destroying these deposits. We investigated the inflammatory response and phagocytic mechanisms when macrophages were exposed to human kidney stones and inorganic crystals. MATERIALS AND METHODS Human monocytes were differentiated into resting, fully differentiated macrophages by treatment with recombinant human macrophage colony-stimulating factor (M-CSF) or GM-CSF (granulocyte M-CSF) for 6 days. After confirming phenotype by flow cytometry the macrophages were exposed for 20 hours to fragments of sterile human calcium oxalate stones or calcium oxalate crystals. Crystal uptake was determined, and supernatant cytokine and chemokine profiles were analyzed using antibody arrays. Quantitative reverse transcriptase-polymerase chain reaction was done to validate mRNA profile expression. RESULTS Under direct vision fluorescence microscopy activated human macrophages were noted to surround stone fragments and synthesized crystals, and destroy them in a step-by-step process that involved clathrin mediated endocytosis and phagocytosis. An inflammatory cascade was released by macrophages, including the chemokines chemokine ligand (CCL)2, CCL3, interleukin (IL)-1 receptor antagonist (IL-1ra), complement component C5/C5a and IL-8. Response patterns to stone and crystal material depended on macrophage phenotype and activation status. CONCLUSIONS In our in vitro study macrophages differentiated with M-CSF showed greater ability to phagocytize crystal deposits than those treated with GM-CSF. Following clathrin mediated endocytosis macrophages released a number of cytokines that are crucial for the inflammatory immune response. This suggests that tissue macrophages have an important role in preventing kidney stone disease by removing and digesting interstitial renal crystal deposits.
Collapse
Affiliation(s)
- Sergei Kusmartsev
- Department of Urology, College of Medicine, University of Florida, Gainesville, Florida
| | | | - Benjamin K Canales
- Department of Urology, College of Medicine, University of Florida, Gainesville, Florida
| | - Vincent G Bird
- Department of Urology, College of Medicine, University of Florida, Gainesville, Florida
| | - Johannes Vieweg
- Department of Urology, College of Medicine, University of Florida, Gainesville, Florida
| | - Saeed R Khan
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, Florida.
| |
Collapse
|
345
|
Niemi JP, DeFrancesco-Lisowitz A, Cregg JM, Howarth M, Zigmond RE. Overexpression of the monocyte chemokine CCL2 in dorsal root ganglion neurons causes a conditioning-like increase in neurite outgrowth and does so via a STAT3 dependent mechanism. Exp Neurol 2015; 275 Pt 1:25-37. [PMID: 26431741 DOI: 10.1016/j.expneurol.2015.09.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 09/08/2015] [Accepted: 09/27/2015] [Indexed: 12/24/2022]
Abstract
Neuroinflammation plays a critical role in the regeneration of peripheral nerves following axotomy. An injury to the sciatic nerve leads to significant macrophage accumulation in the L5 DRG, an effect not seen when the dorsal root is injured. We recently demonstrated that this accumulation around axotomized cell bodies is necessary for a peripheral conditioning lesion response to occur. Here we asked whether overexpression of the monocyte chemokine CCL2 specifically in DRG neurons of uninjured mice is sufficient to cause macrophage accumulation and to enhance regeneration or whether other injury-derived signals are required. AAV5-EF1α-CCL2 was injected intrathecally, and this injection led to a time-dependent increase in CCL2 mRNA expression and macrophage accumulation in L5 DRG, with a maximal response at 3 weeks post-injection. These changes led to a conditioning-like increase in neurite outgrowth in DRG explant and dissociated cell cultures. This increase in regeneration was dependent upon CCL2 acting through its primary receptor CCR2. When CCL2 was overexpressed in CCR2-/- mice, macrophage accumulation and enhanced regeneration were not observed. To address the mechanism by which CCL2 overexpression enhances regeneration, we tested for elevated expression of regeneration-associated genes in these animals. Surprisingly, we found that CCL2 overexpression led to a selective increase in LIF mRNA and neuronal phosphorylated STAT3 (pSTAT3) in L5 DRGs, with no change in expression seen in other RAGs such as GAP-43. Blockade of STAT3 phosphorylation by each of two different inhibitors prevented the increase in neurite outgrowth. Thus, CCL2 overexpression is sufficient to induce macrophage accumulation in uninjured L5 DRGs and increase the regenerative capacity of DRG neurons via a STAT3-dependent mechanism.
Collapse
Affiliation(s)
- Jon P Niemi
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | | | - Jared M Cregg
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Madeline Howarth
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106-4975, USA
| | - Richard E Zigmond
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106-4975, USA.
| |
Collapse
|
346
|
Role of macrophages in Wallerian degeneration and axonal regeneration after peripheral nerve injury. Acta Neuropathol 2015; 130:605-18. [PMID: 26419777 DOI: 10.1007/s00401-015-1482-4] [Citation(s) in RCA: 349] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/22/2015] [Accepted: 09/24/2015] [Indexed: 01/08/2023]
Abstract
The peripheral nervous system (PNS) has remarkable regenerative abilities after injury. Successful PNS regeneration relies on both injured axons and non-neuronal cells, including Schwann cells and immune cells. Macrophages are the most notable immune cells that play key roles in PNS injury and repair. Upon peripheral nerve injury, a large number of macrophages are accumulated at the injury sites, where they not only contribute to Wallerian degeneration, but also are educated by the local microenvironment and polarized to an anti-inflammatory phenotype (M2), thus contributing to axonal regeneration. Significant progress has been made in understanding how macrophages are educated and polarized in the injured microenvironment as well as how they contribute to axonal regeneration. Following the discussion on the main properties of macrophages and their phenotypes, in this review, we will summarize the current knowledge regarding the mechanisms of macrophage infiltration after PNS injury. Moreover, we will discuss the recent findings elucidating how macrophages are polarized to M2 phenotype in the injured PNS microenvironment, as well as the role and underlying mechanisms of macrophages in peripheral nerve injury, Wallerian degeneration and regeneration. Furthermore, we will highlight the potential application by targeting macrophages in treating peripheral nerve injury and peripheral neuropathies.
Collapse
|
347
|
Jurberg AD, Vasconcelos-Fontes L, Cotta-de-Almeida V. A Tale from TGF-β Superfamily for Thymus Ontogeny and Function. Front Immunol 2015; 6:442. [PMID: 26441956 PMCID: PMC4564722 DOI: 10.3389/fimmu.2015.00442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 08/14/2015] [Indexed: 12/16/2022] Open
Abstract
Multiple signaling pathways control every aspect of cell behavior, organ formation, and tissue homeostasis throughout the lifespan of any individual. This review takes an ontogenetic view focused on the large superfamily of TGF-β/bone morphogenetic protein ligands to address thymus morphogenesis and function in T cell differentiation. Recent findings on a role of GDF11 for reversing aging-related phenotypes are also discussed.
Collapse
Affiliation(s)
- Arnon Dias Jurberg
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil ; Graduate Program in Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Larissa Vasconcelos-Fontes
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil
| | - Vinícius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil
| |
Collapse
|
348
|
Protection and pathology in TB: learning from the zebrafish model. Semin Immunopathol 2015; 38:261-73. [PMID: 26324465 PMCID: PMC4779130 DOI: 10.1007/s00281-015-0522-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/11/2015] [Indexed: 12/14/2022]
Abstract
Zebrafish has earned its place among animal models of tuberculosis. Its natural pathogen, Mycobacterium marinum, shares major virulence factors with the human pathogen Mycobacterium tuberculosis. In adult zebrafish, which possess recombination-activated adaptive immunity, it can cause acute infection or a chronic progressive disease with containment of mycobacteria in well-structured, caseating granulomas. In addition, a low-dose model that closely mimics human latent infection has recently been developed. These models are used alongside infection of optically transparent zebrafish embryos and larvae that rely on innate immunity and permit non-invasive visualization of the early stages of developing granulomas that are inaccessible in other animal models. By microinjecting mycobacteria intravenously or into different tissues, systemic and localized infections can be induced, each useful for studying particular aspects of early pathogenesis, such as phagocyte recruitment, granuloma expansion and maintenance, vascularization of granulomas, and the phagocyte-mediated dissemination of mycobacteria. This has contributed to new insights into the mycobacteria-driven mechanisms that promote granuloma formation, the double-edged role of inflammation, the mechanisms of macrophage cell death that favor disease progression, and the host-protective role of autophagy. As a result, zebrafish models are now increasingly used to explore strategies for adjunctive therapy of tuberculosis with host-directed drugs.
Collapse
|
349
|
DeFrancesco-Lisowitz A, Lindborg JA, Niemi JP, Zigmond RE. The neuroimmunology of degeneration and regeneration in the peripheral nervous system. Neuroscience 2015; 302:174-203. [PMID: 25242643 PMCID: PMC4366367 DOI: 10.1016/j.neuroscience.2014.09.027] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 09/08/2014] [Accepted: 09/10/2014] [Indexed: 12/25/2022]
Abstract
Peripheral nerves regenerate following injury due to the effective activation of the intrinsic growth capacity of the neurons and the formation of a permissive pathway for outgrowth due to Wallerian degeneration (WD). WD and subsequent regeneration are significantly influenced by various immune cells and the cytokines they secrete. Although macrophages have long been known to play a vital role in the degenerative process, recent work has pointed to their importance in influencing the regenerative capacity of peripheral neurons. In this review, we focus on the various immune cells, cytokines, and chemokines that make regeneration possible in the peripheral nervous system, with specific attention placed on the role macrophages play in this process.
Collapse
Affiliation(s)
| | - J A Lindborg
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - J P Niemi
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| | - R E Zigmond
- Department of Neurosciences, Case Western Reserve University, Cleveland OH 44106-4975
| |
Collapse
|
350
|
Ji J, Eggert T, Budhu A, Forgues M, Takai A, Dang H, Ye Q, Lee JS, Kim JH, Greten TF, Wang XW. Hepatic stellate cell and monocyte interaction contributes to poor prognosis in hepatocellular carcinoma. Hepatology 2015; 62:481-95. [PMID: 25833323 PMCID: PMC4515211 DOI: 10.1002/hep.27822] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 03/28/2015] [Indexed: 12/14/2022]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) patients suffer from a poor survival rate and a high incidence of postoperative recurrence. The hepatic microenvironment plays a significant role in the initiation, progression, and recurrence of HCC; however, the causal mechanisms of these phenomena are unclear. Given the predominant underlying fibrotic and cirrhotic conditions of the liver prone to HCC and its recurrence, alterations of components of the inflammatory milieu have been suggested as factors that promote HCC development. In particular, activated hepatic stellate cells (A-HSCs), which play a key role in liver fibrosis and cirrhosis, have been suggested as contributors to the HCC-prone microenvironment. Here, we have identified and validated an A-HSC-specific gene expression signature among nontumor tissues of 319 HCC patients that is significantly and independently associated with HCC recurrence and survival. Peritumoral, rather than tumor tissue-related, A-HSC-specific gene expression is associated with recurrence and poor survival. Analyses of A-HSC-specific gene signatures and further immunohistochemical validation in an additional 143 HCC patients have revealed that A-HSCs preferentially affect monocyte populations, shifting their gene expression from an inflammatory to an immunosuppressive signature. In addition, the interaction between A-HSCs and monocytes induces protumorigenic and progressive features of HCC cells by enhancing cell migration and tumor sphere formation. CONCLUSION A-HSCs play a significant role in promoting HCC progression through interaction with and alteration of monocyte activities within the liver microenvironment; thus, disrupting the interactions and signaling events between the inflammatory milieu and components of the microenvironment may be useful therapeutic strategies for preventing HCC tumor relapse.
Collapse
Affiliation(s)
- Juling Ji
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland,Department of Pathology, Medical School of Nantong University, Nantong, China
| | - Tobias Eggert
- Gastrointestinal Malignancy Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Anuradha Budhu
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Marshonna Forgues
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Atsushi Takai
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Hien Dang
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Qinghai Ye
- Liver Cancer Institute, Fudan University, Shanghai, China
| | - Ju-Seog Lee
- Department of Systems Biology, University of Texas, M. D. Anderson Cancer Center, Houston, Texas
| | - Ji Hoon Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Tim F. Greten
- Gastrointestinal Malignancy Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Xin Wei Wang
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland,Corresponding Author: National Cancer Institute, 37 Convent Dr., Bldg. 37, Rm. 3044A, Bethesda, MD 20892;
| |
Collapse
|