501
|
The Reactive Oxygen Species in Macrophage Polarization: Reflecting Its Dual Role in Progression and Treatment of Human Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:2795090. [PMID: 27143992 PMCID: PMC4837277 DOI: 10.1155/2016/2795090] [Citation(s) in RCA: 384] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 03/13/2016] [Accepted: 03/15/2016] [Indexed: 12/18/2022]
Abstract
High heterogeneity of macrophage is associated with its functions in polarization to different functional phenotypes depending on environmental cues. Macrophages remain in balanced state in healthy subject and thus macrophage polarization may be crucial in determining the tissue fate. The two distinct populations, classically M1 and alternatively M2 activated, representing the opposing ends of the full activation spectrum, have been extensively studied for their associations with several disease progressions. Accumulating evidences have postulated that the redox signalling has implication in macrophage polarization and the key roles of M1 and M2 macrophages in tissue environment have provided the clue for the reasons of ROS abundance in certain phenotype. M1 macrophages majorly clearing the pathogens and ROS may be crucial for the regulation of M1 phenotype, whereas M2 macrophages resolve inflammation which favours oxidative metabolism. Therefore how ROS play its role in maintaining the homeostatic functions of macrophage and in particular macrophage polarization will be reviewed here. We also review the biology of macrophage polarization and the disturbance of M1/M2 balance in human diseases. The potential therapeutic opportunities targeting ROS will also be discussed, hoping to provide insights for development of target-specific delivery system or immunomodulatory antioxidant for the treatment of ROS-related diseases.
Collapse
|
502
|
Bosisio FM, Wilmott JS, Volders N, Mercier M, Wouters J, Stas M, Blokx WA, Massi D, Thompson JF, Scolyer RA, van Baren N, van den Oord JJ. Plasma cells in primary melanoma. Prognostic significance and possible role of IgA. Mod Pathol 2016; 29:347-58. [PMID: 26867783 DOI: 10.1038/modpathol.2016.28] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/13/2015] [Accepted: 12/21/2015] [Indexed: 12/20/2022]
Abstract
Melanoma is not only one of the most immunogenic cancers but also one of the most effective cancers at subverting host immunity. The role of T lymphocytes in tumor immunity has been extensively studied in melanoma, whereas less is known about the importance of B lymphocytes. The effects of plasma cells (PCs), in particular, are still obscure. The aim of this study was to characterize pathological features and clinical outcome of primary cutaneous melanomas associated with PCs. Moreover, we investigated the origins of the melanoma-associated PCs. Finally, we studied the outcome of patients with primary melanomas with PCs. We reviewed 710 melanomas to correlate the presence of PCs with histological prognostic markers. Immunohistochemistry for CD138 and heavy and light chains was performed in primary melanomas (PM) and in loco-regional lymph nodes (LN), both metastatic and not metastatic. In three PM and nine LN with frozen material, VDJ-rearrangement was analyzed by Gene Scan Analysis. Survival analysis was performed on a group of 85 primary melanomas >2 mm in thickness. Forty-one cases (3.7%) showed clusters/sheets of PCs. PC-rich melanomas occurred at an older age and were thicker, more often ulcerated and more mitotically active (P<0.05). PCs were polyclonal and often expressed IgA in addition to IgG. In LN, clusters/sheets of IgA+ PCs were found both in the sinuses and subcapsular areas. Analysis of VDJ-rearrangements showed the IgA to be oligoclonal. Melanomas with clusters/sheets of PCs had a significantly worse survival compared with melanomas without PCs while, interestingly, melanomas with sparse PCs were associated with a better clinical outcome (P=0.002). In conclusion, melanomas with sheets/clusters of PCs are associated with worse prognosis. IgG and IgA are the isotypes predominantly produced by these PCs. IgA oligoclonality suggests an antigen-driven response that facilitates melanoma progression by a hitherto unknown mechanism.
Collapse
Affiliation(s)
- Francesca M Bosisio
- Laboratory of Translational Cell and Tissue Research, University of Leuven, KUL, Leuven, Belgium.,Università Degli Studi di Milano-Bicocca, Milan, Italy
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Nathalie Volders
- Laboratory of Translational Cell and Tissue Research, University of Leuven, KUL, Leuven, Belgium
| | - Marjorie Mercier
- Ludwig Institute for Cancer Research and de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Jasper Wouters
- Laboratory of Translational Cell and Tissue Research, University of Leuven, KUL, Leuven, Belgium
| | - Marguerite Stas
- Department of Surgical Oncology, UZ Gasthuisberg and KU Leuven, Leuven, Belgium
| | | | - Daniela Massi
- Department of Surgery and Translational Medicine, University of Florence, Florence, Italy
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Nicolas van Baren
- Ludwig Institute for Cancer Research and de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Joost J van den Oord
- Laboratory of Translational Cell and Tissue Research, University of Leuven, KUL, Leuven, Belgium
| |
Collapse
|
503
|
Han MS, Barrett T, Brehm MA, Davis RJ. Inflammation Mediated by JNK in Myeloid Cells Promotes the Development of Hepatitis and Hepatocellular Carcinoma. Cell Rep 2016; 15:19-26. [PMID: 27052181 PMCID: PMC4826851 DOI: 10.1016/j.celrep.2016.03.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 01/27/2016] [Accepted: 02/26/2016] [Indexed: 01/20/2023] Open
Abstract
The cJun NH2-terminal kinase (JNK) signaling pathway is required for the development of hepatitis and hepatocellular carcinoma. A role for JNK in liver parenchymal cells has been proposed, but more recent studies have implicated non-parenchymal liver cells as the relevant site of JNK signaling. Here, we tested the hypothesis that myeloid cells mediate this function of JNK. We show that mice with myeloid cell-specific JNK deficiency exhibit reduced hepatic inflammation and suppression of both hepatitis and hepatocellular carcinoma. These data identify myeloid cells as a site of pro-inflammatory signaling by JNK that can promote liver pathology. Targeting myeloid cells with a drug that inhibits JNK may therefore provide therapeutic benefit for the treatment of inflammation-related liver disease.
Collapse
Affiliation(s)
- Myoung Sook Han
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Tamera Barrett
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, Worcester, MA 01605, USA
| | - Michael A Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Howard Hughes Medical Institute, Worcester, MA 01605, USA.
| |
Collapse
|
504
|
Iwanowycz S, Wang J, Altomare D, Hui Y, Fan D. Emodin Bidirectionally Modulates Macrophage Polarization and Epigenetically Regulates Macrophage Memory. J Biol Chem 2016; 291:11491-503. [PMID: 27008857 DOI: 10.1074/jbc.m115.702092] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Indexed: 01/21/2023] Open
Abstract
Macrophages are pleiotropic cells capable of performing a broad spectrum of functions. Macrophage phenotypes are classified along a continuum between the extremes of proinflammatory M1 macrophages and anti-inflammatory M2 macrophages. The seemingly opposing functions of M1 and M2 macrophages must be tightly regulated for an effective and proper response to foreign molecules or damaged tissue. Excessive activation of either M1 or M2 macrophages contributes to the pathology of many diseases. Emodin is a Chinese herb-derived compound and has shown potential to inhibit inflammation in various settings. In this study, we tested the ability of emodin to modulate the macrophage response to both M1 and M2 stimuli. Primary mouse macrophages were stimulated with LPS/IFNγ or IL4 with or without emodin, and the effects of emodin on gene transcription, cell signaling pathways, and histone modifications were examined by a variety of approaches, including microarray, quantitative real-time PCR, Western blotting, chromatin immunoprecipitation, and functional assays. We found that emodin bidirectionally tunes the induction of LPS/IFNγ- and IL4-responsive genes through inhibiting NFκB/IRF5/STAT1 signaling and IRF4/STAT6 signaling, respectively. Thereby, emodin modulates macrophage phagocytosis, migration, and NO production. Furthermore, emodin inhibited the removal of H3K27 trimethylation (H3K27m3) marks and the addition of H3K27 acetylation (H3K27ac) marks on genes required for M1 or M2 polarization of macrophages. In conclusion, our data suggest that emodin is uniquely able to suppress the excessive response of macrophages to both M1 and M2 stimuli and therefore has the potential to restore macrophage homeostasis in various pathologies.
Collapse
Affiliation(s)
- Stephen Iwanowycz
- From the Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina 29209 and
| | - Junfeng Wang
- From the Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina 29209 and
| | - Diego Altomare
- the Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, South Carolina 29208
| | - Yvonne Hui
- From the Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina 29209 and
| | - Daping Fan
- From the Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina 29209 and
| |
Collapse
|
505
|
Linares J, Fernández AB, Feito MJ, Matesanz MC, Sánchez-Salcedo S, Arcos D, Vallet-Regí M, Rojo JM, Portolés MT. Effects of nanocrystalline hydroxyapatites on macrophage polarization. J Mater Chem B 2016; 4:1951-1959. [PMID: 32263072 DOI: 10.1039/c6tb00014b] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Silicon substituted and nanocrystalline hydroxyapatites have attracted the attention of many researchers due to their up-regulation in osteoblast cell metabolism and enhanced bioreactivity, respectively. On the other hand, the biomaterial success or failure depends ultimately on the immune response triggered after its implantation. Macrophages are the main components of the innate immune system with an important role in healing and tissue remodelling due to their remarkable functional plasticity, existing in a whole spectrum of functional populations with varying phenotypic features. The effects of nanocrystalline hydroxyapatite (nano-HA) and nanocrystalline silicon substituted hydroxyapatite (nano-SiHA) on the macrophage populations defined as pro-inflammatory (M1) and reparative (M2) phenotypes have been evaluated in the present study using RAW 264.7 cells and mouse peritoneal macrophages as in vitro models. M1 and M2 macrophage phenotypes were characterized by flow cytometry and confocal microscopy by the expression of CD80 and CD163, known as M1 and M2 markers, respectively. The polarization of primary macrophages towards the M1 or M2 phenotype was induced with the pro-inflammatory stimulus LPS or the anti-inflammatory stimulus IL-10, respectively, evaluating the biomaterial effects under these conditions. Our results show that both nano-HA and nano-SiHA favour the macrophage polarization towards an M2 reparative phenotype, decreasing M1 population and ensuring an appropriate response in the implantation site of these biomaterials designed for bone repair and bone tissue engineering.
Collapse
Affiliation(s)
- J Linares
- Department of Biochemistry and Molecular Biology I/Faculty of Chemistry, Universidad Complutense de Madrid, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
506
|
Huang CB, Alimova Y, Ebersole JL. Macrophage polarization in response to oral commensals and pathogens. Pathog Dis 2016; 74:ftw011. [PMID: 26884502 DOI: 10.1093/femspd/ftw011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2016] [Indexed: 01/03/2023] Open
Abstract
Macrophages have been identified in the periodontium. Data have phenotypically described these cells, demonstrated changes with progressing periodontal disease, and identified their ability to function in antigen-presentation critical for adaptive immune responses to individual oral bacterium. Recent evidence has emphasized an important role for the plasticity of macrophage phenotypes, not only in the resulting function of these cells in various tissues, but also clear differences in the stimulatory signals that result in M1 (classical activation, inflammatory) and M2 (alternative activation/deactivated, immunomodulatory) cells. This investigation hypothesized that the oral pathogens, Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans induce M1-type cells, while oral commensal bacteria primarily elicit macrophage functions consistent with an M2 phenotype. However, we observed that the M1 output from P. gingivalis challenge, showed exaggerated levels of pro-inflammatory cytokines, with a much lower production of chemokines related to T-cell recruitment. This contrasted with A. actinomycetemcomitans infection that increased both the pro-inflammatory cytokines and T-cell chemokines. Thus, it appears that P. gingivalis, as an oral pathogen, may have a unique capacity to alter the programming of the M1 macrophage resulting in a hyperinflammatory environment and minimizing the ability for T-cell immunomodulatory influx into the lesions.
Collapse
Affiliation(s)
- Chifu B Huang
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY 40536, USA
| | - Yelena Alimova
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY 40536, USA
| | - Jeffrey L Ebersole
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
507
|
Regulatory effect of moxibustion for rats with ulcerative colitis on the macrophage functional phenotype protein of lung tissue. JOURNAL OF ACUPUNCTURE AND TUINA SCIENCE 2016. [DOI: 10.1007/s11726-016-0891-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
508
|
Zhao JL, Huang F, He F, Gao CC, Liang SQ, Ma PF, Dong GY, Han H, Qin HY. Forced Activation of Notch in Macrophages Represses Tumor Growth by Upregulating miR-125a and Disabling Tumor-Associated Macrophages. Cancer Res 2016; 76:1403-15. [PMID: 26759236 DOI: 10.1158/0008-5472.can-15-2019] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 01/04/2016] [Indexed: 11/16/2022]
Abstract
Tumor-associated macrophages (TAM) contribute greatly to hallmarks of cancer. Notch blockade was shown to arrest TAM differentiation, but the precise role and underlying mechanisms require elucidation. In this study, we employed a transgenic mouse model in which the Notch1 intracellular domain (NIC) is activated conditionally to define the effects of active Notch1 signaling in macrophages. NIC overexpression had no effect on TAM differentiation, but it abrogated TAM function, leading to repressed growth of transplanted tumors. Macrophage miRNA profiling identified a novel downstream mediator of Notch signaling, miR-125a, which was upregulated through an RBP-J-binding site at the first intronic enhancer of the host gene Spaca6A. miR-125a functioned downstream of Notch signaling to reciprocally influence polarization of M1 and M2 macrophages by regulating factor inhibiting hypoxia inducible factor-1α and IRF4, respectively. Notably, macrophages transfected with miR-125a mimetics increased phagocytic activity and repressed tumor growth by remodeling the immune microenvironment. We also identified a positive feedback loop for miR-125a expression mediated by RYBP and YY1. Taken together, our results showed that Notch signaling not only supported the differentiation of TAM but also antagonized their protumorigenic function through miR-125a. Targeting this miRNA may reprogram macrophages in the tumor microenvironment and restore their antitumor potential.
Collapse
Affiliation(s)
- Jun-Long Zhao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Fei Huang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Fei He
- Department of Hepatic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chun-Chen Gao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Shi-Qian Liang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Peng-Fei Ma
- Department of Hepatic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guang-Ying Dong
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China. Department of Hepatic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Hong-Yan Qin
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
509
|
Watanabe N, Suzuki Y, Inokuchi S, Inoue S. Sepsis induces incomplete M2 phenotype polarization in peritoneal exudate cells in mice. J Intensive Care 2016; 4:6. [PMID: 26759721 PMCID: PMC4709882 DOI: 10.1186/s40560-015-0124-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 12/29/2015] [Indexed: 11/10/2022] Open
Abstract
Background Macrophages can differentiate into pro-inflammatory (M1) or anti-inflammatory (M2) phenotypes upon exposure to a pathogen or a cytokine microenvironment. However, M1/M2 macrophage polarization in polymicrobial sepsis has not been fully characterized. Methods The polarity of peritoneal exudate (PE) cells from mice that had undergone cecal ligation and puncture (CLP) and the response of those cells to lipopolysaccharide (LPS) in terms of cytokine and chemokine expression were examined. Results PE cells from CLP mice demonstrated a shift toward the M2 phenotype in terms of marker enzyme expression. In addition, the CLP-derived PE cells showed apparent unresponsiveness to LPS stimulation with regard to expression of pro-inflammatory cytokines such as TNF-α, while the expression of anti-inflammatory cytokines such as IL-10 was induced. Nevertheless, the CLP-PE cells failed to express M2 chemokines including chemokine (C-C motif) ligand 17 (CCL17), CCL22, and CCL24, all of which are important for T cell recruitment. Conclusions The results suggested that a shift of naïve monocytes/macrophages to the M2 phenotype, along with the lack of M2 chemokine expression in septic monocytes/macrophages, might be responsible for immunosuppression after sepsis. Electronic supplementary material The online version of this article (doi:10.1186/s40560-015-0124-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nobuo Watanabe
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Shimokasuya 143, Isehara, Kanagawa 259-1193 Japan
| | - Yusuke Suzuki
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Shimokasuya 143, Isehara, Kanagawa 259-1193 Japan
| | - Sadaki Inokuchi
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Shimokasuya 143, Isehara, Kanagawa 259-1193 Japan
| | - Shigeaki Inoue
- Department of Emergency and Critical Care Medicine, Tokai University School of Medicine, Shimokasuya 143, Isehara, Kanagawa 259-1193 Japan
| |
Collapse
|
510
|
Zheng Z, Zheng F. Immune Cells and Inflammation in Diabetic Nephropathy. J Diabetes Res 2016; 2016:1841690. [PMID: 26824038 PMCID: PMC4707326 DOI: 10.1155/2016/1841690] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 10/21/2015] [Indexed: 12/20/2022] Open
Abstract
Diabetic nephropathy (DN) is a serious complication of diabetes. At its core, DN is a metabolic disorder which can also manifest itself in terms of local inflammation in the kidneys. Such inflammation can then drive the classical markers of fibrosis and structural remodeling. As a result, resolution of immune-mediated inflammation is critical towards achieving a cure for DN. Many immune cells play a part in DN, including key members of both the innate and adaptive immune systems. While these cells were classically understood to primarily function against pathogen insult, it has also become increasingly clear that they also serve a major role as internal sensors of damage. In fact, damage sensing may serve as the impetus for much of the inflammation that occurs in DN, in a vicious positive feedback cycle. Although direct targeting of these proinflammatory cells may be difficult, new approaches that focus on their metabolic profiles may be able to alleviate DN significantly, especially since dysregulation of the local metabolic environment may well be responsible for triggering inflammation to begin with. In this review, the authors consider the metabolic profile of several relevant immune types and discuss their respective roles.
Collapse
Affiliation(s)
- Zihan Zheng
- College of Arts and Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Feng Zheng
- Department of Nephrology, Advanced Institute for Medical Sciences, Second Hospital, Dalian Medical University, Dalian 116023, China
- Department of Nephrology and Basic Science Laboratory, Fujian Medical University, Fuzhou 350002, China
- *Feng Zheng:
| |
Collapse
|
511
|
Salmiheimo ANE, Mustonen HK, Vainionpää SAA, Shen Z, Kemppainen EAJ, Seppänen HE, Puolakkainen PA. Increasing the Inflammatory Competence of Macrophages with IL-6 or with Combination of IL-4 and LPS Restrains the Invasiveness of Pancreatic Cancer Cells. J Cancer 2016; 7:42-9. [PMID: 26722359 PMCID: PMC4679380 DOI: 10.7150/jca.12923] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022] Open
Abstract
Recent studies suggest that pro-inflammatory type M1 macrophages inhibit tumor progression and that anti-inflammatory M2 macrophages enhance it. The aim of this study was to examine the interaction of type M1 and M2 macrophages with pancreatic cancer cells. We studied the migration rate of fluorescein stained pancreatic cancer cells on Matrigel cultured alone or with Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) differentiated macrophages or with Macrophage Colony Stimulating Factor (M-CSF) differentiated macrophages, skewing the phenotype towards pro- and anti-inflammatory direction, respectively. Macrophage differentiation was assessed with flow cytometry and the cytokine secretion in cell cultures with cytokine array. Both GM-CSF and M-CSF differentiated macrophages increased the migration rate of primary pancreatic adenocarcinoma cell line (MiaPaCa-2) and metastatic cell line (HPAF-II). Stimulation with IL6 or IL4+LPS reversed the macrophages' increasing effect on the migration rate of MiaPaCa-2 completely and partly of HPAF-II. Co-culture with MiaPaCa-2 reduced the inflammatory cytokine secretion of GM-CSF differentiated macrophages. Co-culture of macrophages with pancreatic cancer cells seem to change the inflammatory cytokine profile of GM-CSF differentiated macrophages and this might explain why also GM-CSF differentiated macrophages promoted the invasion. Adding IL6 or IL4+LPS to the cell culture with MiaPaCa-2 and GM-CSF or M-CSF differentiated macrophages increased the secretion of inflammatory cytokines and this could contribute to the reversion of the macrophage induced increase of cancer cell migration rate.
Collapse
Affiliation(s)
- Aino N E Salmiheimo
- 1. Department of Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Harri K Mustonen
- 1. Department of Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Sanna A A Vainionpää
- 1. Department of Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Zhanlong Shen
- 2. Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing, China
| | - Esko A J Kemppainen
- 1. Department of Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Hanna E Seppänen
- 1. Department of Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Pauli A Puolakkainen
- 1. Department of Surgery, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
512
|
Jiang J, Jia T, Gong W, Ning B, Wooley PH, Yang SY. Macrophage Polarization in IL-10 Treatment of Particle-Induced Inflammation and Osteolysis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:57-66. [DOI: 10.1016/j.ajpath.2015.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/14/2015] [Accepted: 09/18/2015] [Indexed: 01/11/2023]
|
513
|
Bennett BJ, Davis RC, Civelek M, Orozco L, Wu J, Qi H, Pan C, Packard RRS, Eskin E, Yan M, Kirchgessner T, Wang Z, Li X, Gregory JC, Hazen SL, Gargalovic PS, Lusis AJ. Genetic Architecture of Atherosclerosis in Mice: A Systems Genetics Analysis of Common Inbred Strains. PLoS Genet 2015; 11:e1005711. [PMID: 26694027 PMCID: PMC4687930 DOI: 10.1371/journal.pgen.1005711] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 11/06/2015] [Indexed: 12/15/2022] Open
Abstract
Common forms of atherosclerosis involve multiple genetic and environmental factors. While human genome-wide association studies have identified numerous loci contributing to coronary artery disease and its risk factors, these studies are unable to control environmental factors or examine detailed molecular traits in relevant tissues. We now report a study of natural variations contributing to atherosclerosis and related traits in over 100 inbred strains of mice from the Hybrid Mouse Diversity Panel (HMDP). The mice were made hyperlipidemic by transgenic expression of human apolipoprotein E-Leiden (APOE-Leiden) and human cholesteryl ester transfer protein (CETP). The mice were examined for lesion size and morphology as well as plasma lipid, insulin and glucose levels, and blood cell profiles. A subset of mice was studied for plasma levels of metabolites and cytokines. We also measured global transcript levels in aorta and liver. Finally, the uptake of acetylated LDL by macrophages from HMDP mice was quantitatively examined. Loci contributing to the traits were mapped using association analysis, and relationships among traits were examined using correlation and statistical modeling. A number of conclusions emerged. First, relationships among atherosclerosis and the risk factors in mice resemble those found in humans. Second, a number of trait-loci were identified, including some overlapping with previous human and mouse studies. Third, gene expression data enabled enrichment analysis of pathways contributing to atherosclerosis and prioritization of candidate genes at associated loci in both mice and humans. Fourth, the data provided a number of mechanistic inferences; for example, we detected no association between macrophage uptake of acetylated LDL and atherosclerosis. Fifth, broad sense heritability for atherosclerosis was much larger than narrow sense heritability, indicating an important role for gene-by-gene interactions. Sixth, stepwise linear regression showed that the combined variations in plasma metabolites, including LDL/VLDL-cholesterol, trimethylamine N-oxide (TMAO), arginine, glucose and insulin, account for approximately 30 to 40% of the variation in atherosclerotic lesion area. Overall, our data provide a rich resource for studies of complex interactions underlying atherosclerosis. While recent genetic association studies in human populations have succeeded in identifying genetic loci that contribute to coronary artery disease (CAD) and related phenotypes, these loci explain only a small fraction of the genetic variation in CAD and associated traits. Here, we present a complementary approach using association analysis of atherosclerotic traits among inbred strains of mice. A strength of this approach is that it enables in-depth phenotypic characterization including gene expression and metabolic profiling across a variety of tissues, and integration of these molecular phenotypes with coronary artery disease itself. A striking finding was the large fraction of atherosclerosis that was explained by genetic interactions. Association analysis allowed us to identify genetic loci for atherosclerotic lesion area as well as transcript, cytokine and metabolite levels, and relationships among the traits were examined by correlation and network modeling. The plasma metabolites associated with atherosclerosis in mice, namely, LDL/VLDL-cholesterol, TMAO, arginine, glucose and insulin, overlapped with those observed in humans and accounted for approximately 30 to 40% of the observed variation in atherosclerotic lesion area. In summary, our data provide a detailed overview of the genetic architecture of atherosclerosis in mice and a rich resource for studies of the complex genetic and metabolic interactions that underlie the disease.
Collapse
Affiliation(s)
- Brian J. Bennett
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Richard C. Davis
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Mete Civelek
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Luz Orozco
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Judy Wu
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Hannah Qi
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Calvin Pan
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - René R. Sevag Packard
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Eleazar Eskin
- Department of Computer Science, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Mujing Yan
- Department of Cardiovascular Drug Discovery, Bristol-Myers Squibb, Princeton, New Jersey, United States of America
| | - Todd Kirchgessner
- Department of Cardiovascular Drug Discovery, Bristol-Myers Squibb, Princeton, New Jersey, United States of America
| | - Zeneng Wang
- Department of Cellular and Molecular Medicine (NC10), Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States of America
| | - Xinmin Li
- Department of Cellular and Molecular Medicine (NC10), Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States of America
| | - Jill C. Gregory
- Department of Cellular and Molecular Medicine (NC10), Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States of America
| | - Stanley L. Hazen
- Department of Cellular and Molecular Medicine (NC10), Cleveland Clinic Lerner Research Institute, Cleveland, Ohio, United States of America
| | - Peter S. Gargalovic
- Department of Cardiovascular Drug Discovery, Bristol-Myers Squibb, Princeton, New Jersey, United States of America
| | - Aldons J. Lusis
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
514
|
Geniposide alleviates inflammation by suppressing MeCP2 in mice with carbon tetrachloride-induced acute liver injury and LPS-treated THP-1 cells. Int Immunopharmacol 2015; 29:739-747. [DOI: 10.1016/j.intimp.2015.08.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/18/2015] [Accepted: 08/31/2015] [Indexed: 12/29/2022]
|
515
|
Casey SC, Amedei A, Aquilano K, Azmi AS, Benencia F, Bhakta D, Bilsland AE, Boosani CS, Chen S, Ciriolo MR, Crawford S, Fujii H, Georgakilas AG, Guha G, Halicka D, Helferich WG, Heneberg P, Honoki K, Keith WN, Kerkar SP, Mohammed SI, Niccolai E, Nowsheen S, Vasantha Rupasinghe HP, Samadi A, Singh N, Talib WH, Venkateswaran V, Whelan RL, Yang X, Felsher DW. Cancer prevention and therapy through the modulation of the tumor microenvironment. Semin Cancer Biol 2015; 35 Suppl:S199-S223. [PMID: 25865775 PMCID: PMC4930000 DOI: 10.1016/j.semcancer.2015.02.007] [Citation(s) in RCA: 255] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 02/06/2023]
Abstract
Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.
Collapse
Affiliation(s)
- Stephanie C Casey
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Asfar S Azmi
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Fabian Benencia
- Department of Biomedical Sciences, Ohio University, Athens, OH, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | - Alan E Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Chandra S Boosani
- Department of Biomedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Sophie Chen
- Ovarian and Prostate Cancer Research Laboratory, Guildford, Surrey, United Kingdom
| | | | - Sarah Crawford
- Department of Biology, Southern Connecticut State University, New Haven, CT, United States
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur 613401, Tamil Nadu, India
| | | | - William G Helferich
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Petr Heneberg
- Charles University in Prague, Third Faculty of Medicine, Prague, Czech Republic
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Japan
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sid P Kerkar
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | | | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture, Dalhousie University, Nova Scotia, Canada
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science University, Amman, Jordan
| | | | - Richard L Whelan
- Mount Sinai Roosevelt Hospital, Icahn Mount Sinai School of Medicine, New York City, NY, United States
| | - Xujuan Yang
- University of Illinois at Urbana-Champaign, Champaign-Urbana, IL, United States
| | - Dean W Felsher
- Division of Oncology, Departments of Medicine and Pathology, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
516
|
Hodgkinson JW, Grayfer L, Belosevic M. Biology of Bony Fish Macrophages. BIOLOGY 2015; 4:881-906. [PMID: 26633534 PMCID: PMC4690021 DOI: 10.3390/biology4040881] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 11/20/2015] [Accepted: 11/24/2015] [Indexed: 01/21/2023]
Abstract
Macrophages are found across all vertebrate species, reside in virtually all animal tissues, and play critical roles in host protection and homeostasis. Various mechanisms determine and regulate the highly plastic functional phenotypes of macrophages, including antimicrobial host defenses (pro-inflammatory, M1-type), and resolution and repair functions (anti-inflammatory/regulatory, M2-type). The study of inflammatory macrophages in immune defense of teleosts has garnered much attention, and antimicrobial mechanisms of these cells have been extensively studied in various fish models. Intriguingly, both similarities and differences have been documented for the regulation of lower vertebrate macrophage antimicrobial defenses, as compared to what has been described in mammals. Advances in our understanding of the teleost macrophage M2 phenotypes likewise suggest functional conservation through similar and distinct regulatory strategies, compared to their mammalian counterparts. In this review, we discuss the current understanding of the molecular mechanisms governing teleost macrophage functional heterogeneity, including monopoetic development, classical macrophage inflammatory and antimicrobial responses as well as alternative macrophage polarization towards tissues repair and resolution of inflammation.
Collapse
Affiliation(s)
- Jordan W Hodgkinson
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada.
| | - Leon Grayfer
- Department of Biological Sciences, George Washington University, Washington, DC 20052, USA.
| | - Miodrag Belosevic
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada.
| |
Collapse
|
517
|
Flannagan RS, Heit B, Heinrichs DE. Antimicrobial Mechanisms of Macrophages and the Immune Evasion Strategies of Staphylococcus aureus. Pathogens 2015; 4:826-68. [PMID: 26633519 PMCID: PMC4693167 DOI: 10.3390/pathogens4040826] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 11/17/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022] Open
Abstract
Habitually professional phagocytes, including macrophages, eradicate microbial invaders from the human body without overt signs of infection. Despite this, there exist select bacteria that are professional pathogens, causing significant morbidity and mortality across the globe and Staphylococcus aureus is no exception. S. aureus is a highly successful pathogen that can infect virtually every tissue that comprises the human body causing a broad spectrum of diseases. The profound pathogenic capacity of S. aureus can be attributed, in part, to its ability to elaborate a profusion of bacterial effectors that circumvent host immunity. Macrophages are important professional phagocytes that contribute to both the innate and adaptive immune response, however from in vitro and in vivo studies, it is evident that they fail to eradicate S. aureus. This review provides an overview of the antimicrobial mechanisms employed by macrophages to combat bacteria and describes the immune evasion strategies and some representative effectors that enable S. aureus to evade macrophage-mediated killing.
Collapse
Affiliation(s)
- Ronald S Flannagan
- Department of Microbiology and Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
| | - Bryan Heit
- Department of Microbiology and Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
- Centre for Human Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
| | - David E Heinrichs
- Department of Microbiology and Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
- Centre for Human Immunology, the University of Western Ontario, London, ON N6A 5C1, Canada.
| |
Collapse
|
518
|
Belcastro E, Franzini M, Cianchetti S, Lorenzini E, Masotti S, Fierabracci V, Pucci A, Pompella A, Corti A. Monocytes/macrophages activation contributes to b-gamma-glutamyltransferase accumulation inside atherosclerotic plaques. J Transl Med 2015; 13:325. [PMID: 26463174 PMCID: PMC4604102 DOI: 10.1186/s12967-015-0687-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 10/04/2015] [Indexed: 11/30/2022] Open
Abstract
Background Gamma-glutamyltransferase (GGT) is a well-established independent risk factor for cardiovascular mortality related to atherosclerotic disease. Four GGT fractions have been identified in plasma, but only b-GGT fraction accumulates in atherosclerotic plaques, and correlates with other histological markers of vulnerability. The present study was aimed to evaluate whether macrophagic lineage cells may provide a source of b-GGT within the atherosclerotic plaque. Methods GGT expression and release were studied in human monocytes isolated from peripheral blood of healthy donors. The growth factors GM-CSF and M-CSF were used to induce differentiation into M1-like and M2-like macrophages, respectively. Plaque GGT was investigated in tissue samples obtained from patients undergoing carotid endoarterectomy. Results We found that M1-like macrophages express higher levels of GGT as compared to M2-like, and that both monocytes and M1-like macrophages—but not M2-like—are able to release the b-GGT fraction upon activation with pro-inflammatory stimuli. Western blot analysis of b-GGT extracted from plaques confirmed the presence of a GGT immunoreactive peptide coincident with that of macrophages. Conclusions Our data indicate that macrophages characterized by a pro-inflammatory phenotype may contribute to intra-plaque accumulation of b-GGT, which in turn may play a role in the progression of atherosclerosis by modulating inflammatory processes and favouring plaque instability.
Collapse
Affiliation(s)
- Eugenia Belcastro
- Department of Translational Research and New Technologies in Medicine and Surgery, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy. .,CITHEFOR-EA 3452, Faculté de Pharmacie, Université de Lorraine, Nancy, France.
| | - Maria Franzini
- Department of Translational Research and New Technologies in Medicine and Surgery, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Silvana Cianchetti
- Department of Surgery and Medical, Molecular, and Critical Area Pathology, Medical School, University of Pisa, Pisa, Italy.
| | - Evelina Lorenzini
- Department of Translational Research and New Technologies in Medicine and Surgery, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Silvia Masotti
- Life Science Institute, Scuola Superiore Sant'Anna, Pisa, Italy.
| | - Vanna Fierabracci
- Department of Translational Research and New Technologies in Medicine and Surgery, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Angela Pucci
- Histopathology Department, University Hospital, Pisa, Italy.
| | - Alfonso Pompella
- Department of Translational Research and New Technologies in Medicine and Surgery, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Alessandro Corti
- Department of Translational Research and New Technologies in Medicine and Surgery, Medical School, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| |
Collapse
|
519
|
Nie Y, Lu YX, Lv LH. Effect of propofol on generation of inflammatory mediator of monocytes. ASIAN PAC J TROP MED 2015; 8:964-970. [PMID: 26614998 DOI: 10.1016/j.apjtm.2015.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 09/20/2015] [Accepted: 09/30/2015] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVE To evaluate the effect of propofol with different concentrations on the expression of inflammatory mediators of interleukin and tumor-necrosis factor-α (TNF-α) by stimulating the mouse primary monocytes and human monocytic cell line with lipopolysaccharide (LPS) and also discuss the effect of propofol on the secretion of inflammatory mediator and its possible molecular mechanism. METHODS The mononuclear cells of mouse spleen were separated and then purified to obtain the primary monocytes. The dose-effect relationship of production of pro-inflammatory cytokines by monocytes which were stimulated by LPS, namely the monocytes were stimulated by the dose of 0-500 ng/mL for 24 h. ELISA was employed to detect the concentration of IL-6, IL-8 and TNF-α. The effect of propofol on the secretion of above pro-inflammatory cytokines by the monocytes was observed. Cells were divided into the control group, the 0.1% DMSO group, the LPS group and the treatment group with LPS + different dose of propofol (propofol 1-100 μg/mL). ELISA was employed to detect the concentration of IL-6, IL-8 and TNF-α. The change in the expression of important signaling molecules in Toll-like receptor and NF-κB signaling pathway was detected after THP-1 cells were treated with propofol. RESULTS The concentration of TNF-α was (3863 ± 153) pg/mL after 12 h of stimulation by LPS and then its concentration was decreased gradually. But the concentration of IL-6 and IL-8 was relatively high after 24 h of stimulation by LPS, (5627 ± 330) pg/mL and (1626 ± 200) pg/mL, respectively. The propofol could inhibit the expression of IL-6, IL-8 and TNF-α caused by LPS. After the intervention treatment of 50 μg/mL propofol, the concentration of IL-6, IL-8 and TNF-α was significantly decreased (P < 0.01). CONCLUSIONS The propofol can inhibit the expression of TLR-4 and NF-κB to inhibit the activation of p38 and the expression of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Yun Nie
- Operating Room, First People's Hospital of Ji'nan, Shandong 250011, China.
| | - Yan-Xi Lu
- Operating Room, First People's Hospital of Ji'nan, Shandong 250011, China
| | - Li-Hong Lv
- Operating Room, First People's Hospital of Ji'nan, Shandong 250011, China
| |
Collapse
|
520
|
Gonzalez OA, Novak MJ, Kirakodu S, Stromberg A, Nagarajan R, Huang CB, Chen KC, Orraca L, Martinez-Gonzalez J, Ebersole JL. Differential Gene Expression Profiles Reflecting Macrophage Polarization in Aging and Periodontitis Gingival Tissues. Immunol Invest 2015; 44:643-64. [PMID: 26397131 DOI: 10.3109/08820139.2015.1070269] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Recent evidence has determined a phenotypic and functional heterogeneity for macrophage populations. This plasticity of macrophage function has been related to specific properties of subsets (M1 and M2) of these cells in inflammation, adaptive immune responses and resolution of tissue destructive processes. This investigation hypothesized that targeted alterations in the distribution of macrophage phenotypes in aged individuals, and with periodontitis would be skewed towards M1 inflammatory macrophages in gingival tissues. The study used a non-human primate model to evaluate gene expression profiles as footprints of macrophage variation in healthy and periodontitis gingival tissues from animals 3-23 years of age and in periodontitis tissues in adult and aged animals. Significant increases in multiple genes reflecting overall increases in macrophage activities were observed in healthy aged tissues, and were significantly increased in periodontitis tissues from both adults and aged animals. Generally, gene expression patterns for M2 macrophages were similar in healthy young, adolescent and adult tissues. However, modest increases were noted in healthy aged tissues, similar to those seen in periodontitis tissues from both age groups. M1 macrophage gene transcription patterns increased significantly over the age range in healthy tissues, with multiple genes (e.g. CCL13, CCL19, CCR7 and TLR4) significantly increased in aged animals. Additionally, gene expression patterns for M1 macrophages were significantly increased in adult health versus periodontitis and aged healthy versus periodontitis. The findings supported a significant increase in macrophages with aging and in periodontitis. The primary increases in both healthy aged tissues and, particularly periodontitis tissues appeared in the M1 phenotype.
Collapse
Affiliation(s)
- O A Gonzalez
- a Center for Oral Health Research, College of Dentistry, University of Kentucky , Lexington , KY , USA
| | - M J Novak
- a Center for Oral Health Research, College of Dentistry, University of Kentucky , Lexington , KY , USA
| | - S Kirakodu
- a Center for Oral Health Research, College of Dentistry, University of Kentucky , Lexington , KY , USA
| | - A Stromberg
- b Department of Statistics , College of Arts and Sciences, University of Kentucky , Lexington , KY , USA
| | - R Nagarajan
- c Department of Biostatistics , College of Public Health, University of Puerto Rico , San Juan , PR , USA
| | - C B Huang
- a Center for Oral Health Research, College of Dentistry, University of Kentucky , Lexington , KY , USA
| | - K C Chen
- d Microarray Core Facility, College of Medicine, University of Puerto Rico , San Juan , PR , USA
| | - L Orraca
- e School of Dental Medicine, University of Puerto Rico , San Juan , PR , USA , and
| | - J Martinez-Gonzalez
- f Caribbean Primate Research Center, University of Puerto Rico , San Juan , PR , USA
| | - J L Ebersole
- a Center for Oral Health Research, College of Dentistry, University of Kentucky , Lexington , KY , USA
| |
Collapse
|
521
|
Hu Z, Pei G, Wang P, Yang J, Zhu F, Guo Y, Wang M, Yao Y, Zeng R, Liao W, Xu G. Biliverdin Reductase A (BVRA) Mediates Macrophage Expression of Interleukin-10 in Injured Kidney. Int J Mol Sci 2015; 16:22621-35. [PMID: 26393580 PMCID: PMC4613327 DOI: 10.3390/ijms160922621] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/02/2015] [Accepted: 09/08/2015] [Indexed: 12/19/2022] Open
Abstract
Biliverdin reductase A is an enzyme, with serine/threonine/tyrosine kinase activation, converting biliverdin (BV) to bilirubin (BR) in heme degradation pathway. It has been reported to have anti-inflammatory and antioxidant effect in monocytes and human glioblastoma. However, the function of BVRA in polarized macrophage was unknown. This study aimed to investigate the effect of BVRA on macrophage activation and polarization in injured renal microenvironment. Classically activated macrophages (M1macrophages) and alternative activation of macrophages (M2 macrophages) polarization of murine bone marrow derived macrophage was induced by GM-CSF and M-CSF. M1 polarization was associated with a significant down-regulation of BVRA and Interleukin-10 (IL-10), and increased secretion of TNF-α. We also found IL-10 expression was increased in BVRA over-expressed macrophages, while it decreased in BVRA knockdown macrophages. In contrast, BVRA over-expressed or knockdown macrophages had no effect on TNF-α expression level, indicating BVRA mediated IL-10 expression in macrophages. Furthermore, we observed in macrophages infected with recombinant adenoviruses BVRA gene, which BVRA over-expressed enhanced both INOS and ARG-1 mRNA expression, resulting in a specific macrophage phenotype. Through in vivo study, we found BVRA positive macrophages largely existed in mice renal ischemia perfusion injury. With the treatment of the regular cytokines GM-CSF, M-CSF or LPS, excreted in the injured renal microenvironment, IL-10 secretion was significantly increased in BVRA over-expressed macrophages. In conclusion, the BVRA positive macrophage is a source of anti-inflammatory cytokine IL-10 in injured kidney, which may provide a potential target for treatment of kidney disease.
Collapse
Affiliation(s)
- Zhizhi Hu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Guangchang Pei
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Pengge Wang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Juan Yang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Fengmin Zhu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Yujiao Guo
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Meng Wang
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Ying Yao
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Rui Zeng
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Wenhui Liao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| | - Gang Xu
- Division of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Ave, Wuhan 430030, Hubei, China.
| |
Collapse
|
522
|
Fontana MF, Baccarella A, Kellar D, Oniskey TK, Terinate P, Rosenberg SD, Huang EJ, Herbert DR, Kim CC. Myeloid expression of the AP-1 transcription factor JUNB modulates outcomes of type 1 and type 2 parasitic infections. Parasite Immunol 2015; 37:470-8. [PMID: 26178310 PMCID: PMC4573644 DOI: 10.1111/pim.12215] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/03/2015] [Indexed: 12/31/2022]
Abstract
Activation of macrophages is a key step in the initiation of immune responses, but the transcriptional mechanisms governing macrophage activation during infection are not fully understood. It was recently shown that the AP-1 family transcription factor JUNB positively regulates macrophage activation in response to Toll-like receptor agonists that promote classical or M1 polarization, as well as to the cytokine interleukin-4 (IL-4), which elicits an alternatively activated or M2 phenotype. However, a role for JUNB in macrophage activation has never been demonstrated in vivo. Here, to dissect the role of JUNB in macrophage activation in a physiological setting, mice lacking JUNB specifically in myeloid cells were tested in two infection models: experimental cerebral malaria, which elicits a pathological type 1 immune response, and helminth infection, in which type 2 responses are protective. Myeloid-restricted deletion of Junb reduced type 1 immune activation, which was associated with reduced cerebral pathology and improved survival during infection with Plasmodium berghei. Myeloid JUNB deficiency also compromised type 2 activation during infection with the hookworm Nippostrongylus brasiliensis, leading to diminished cytokine production and eosinophil recruitment and increased parasite burden. These results demonstrate that JUNB in myeloid cells shapes host responses and outcomes during type 1 and type 2 infections.
Collapse
Affiliation(s)
- Mary F. Fontana
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alyssa Baccarella
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Danielle Kellar
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Taylor K. Oniskey
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Paul Terinate
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sarah D. Rosenberg
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric J. Huang
- Departments of Pathology and Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - De’Broski R. Herbert
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Charles C. Kim
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
523
|
Current Concept and Update of the Macrophage Plasticity Concept: Intracellular Mechanisms of Reprogramming and M3 Macrophage "Switch" Phenotype. BIOMED RESEARCH INTERNATIONAL 2015; 2015:341308. [PMID: 26366410 PMCID: PMC4561113 DOI: 10.1155/2015/341308] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 07/18/2015] [Accepted: 07/22/2015] [Indexed: 12/17/2022]
Abstract
Macrophages play a key role in immunity. In this review, we consider the traditional notion of macrophage plasticity, data that do not fit into existing concepts, and a hypothesis for existence of a new switch macrophage phenotype. Depending on the microenvironment, macrophages can reprogram their phenotype toward the proinflammatory M1 phenotype or toward the anti-inflammatory M2 phenotype. Macrophage reprogramming involves well-coordinated changes in activities of signalling and posttranslational mechanisms. Macrophage reprogramming is provided by JNK-, PI3K/Akt-, Notch-, JAK/STAT-, TGF-β-, TLR/NF-κB-, and hypoxia-dependent pathways. Posttranscriptional regulation is based on micro-mRNA. We have hypothesized that, in addition to the M1 and M2 phenotypes, an M3 switch phenotype exists. This switch phenotype responds to proinflammatory stimuli with reprogramming towards the anti-inflammatory M2 phenotype or, contrarily, it responds to anti-inflammatory stimuli with reprogramming towards the proinflammatory M1 phenotype. We have found signs of such a switch phenotype in lung diseases. Understanding the mechanisms of macrophage reprogramming will assist in the selection of new therapeutic targets for correction of impaired immunity.
Collapse
|
524
|
Macrophages Contribute to the Progression of Infantile Hemangioma by Regulating the Proliferation and Differentiation of Hemangioma Stem Cells. J Invest Dermatol 2015; 135:3163-3172. [PMID: 26288359 DOI: 10.1038/jid.2015.321] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 07/14/2015] [Accepted: 07/20/2015] [Indexed: 02/06/2023]
Abstract
Macrophage infiltration has been implicated in infantile hemangioma (IH), the most common tumor of infancy. However, the exact role of macrophages in IH remains unknown. This study aims to clarify the functional significance of macrophages in the progression of IH. The distribution of macrophages in human IH was analyzed, and our results revealed that polarized macrophages were more prevalent in proliferating IHs than in involuting IHs, which was consistent with the increased macrophage-related cytokines in proliferating IHs. In vitro results further demonstrated that polarized macrophages effectively promoted the proliferation of hemangioma stem cells (HemSCs) and suppressed their adipogenesis in an Akt- and extracellular signal-regulated kinase 1/2 (Erk1/2)-dependent manner. Moreover, M2- but not M1-polarized macrophages promoted the endothelial differentiation of HemSCs. Furthermore, mixing macrophages in a murine hemangioma model elevated microvessel density and postponed fat tissue formation, which was concomitant with the activation of Akt and Erk1/2 signals. Cluster analysis revealed a close correlation among the macrophage markers, Ki67, vascular endothelial growth factor (VEGF), p-Akt, and p-Erk1/2 in human IH tissues. Collectively, our results suggest that macrophages in IH contribute to tumor progression by promoting the proliferation and endothelial differentiation while suppressing the adipogenesis of HemSCs. These findings indicate that targeting the infiltrating macrophages in IH is a promising therapeutic approach to accelerate IH regression.
Collapse
|
525
|
Kurtzberg J, Buntz S, Gentry T, Noeldner P, Ozamiz A, Rusche B, Storms RW, Wollish A, Wenger DA, Balber AE. Reprint of: Preclinical characterization of DUOC-01, a cell therapy product derived from banked umbilical cord blood for use as an adjuvant to umbilical cord blood transplantation for treatment of inherited metabolic diseases. Cytotherapy 2015; 17:1314-26. [PMID: 26276011 DOI: 10.1016/j.jcyt.2015.07.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 02/09/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND AIMS Cord blood (CB) transplantation slows neurodegeneration during certain inherited metabolic diseases. However, the number of donor cells in the brain of patients does not appear to be sufficient to provide benefit until several months after transplant. We developed the cell product DUOC-01 to provide therapeutic effects in the early post-transplant period. METHODS DUOC-01 cultures initiated from banked CB units were characterized by use of time-lapse photomicroscopy during the 21-day manufacturing process. Antigen expression was measured by means of flow cytometry and immunocytochemistry; transcripts for cytokines and enzymes by quantitative real-time polymerase chain reaction; activities of lysosomal enzymes by direct biochemical analysis; alloreactivity of DUOC-01 and of peripheral blood (PB) mononuclear cells (MNC) to DUOC-01 by mixed lymphocyte culture methods; and cytokine secretion by Bioplex assays. RESULTS DUOC-01 cultures contained highly active, attached, motile, slowly proliferating cells that expressed common (cluster of differentiation [CD]11b, CD14 and Iba1), M1 type (CD16, inducible nitric oxide synthase), and M2-type (CD163, CD206) macrophage or microglia markers. Activities of 11 disease-relevant lysosomal enzymes in DUOC-01 products were similar to those of normal PB cells. All DUOC-01 products secreted interleukin (IL)-6 and IL-10. Accumulation of transforming growth factor-β, IL-1β, interferon-γ and TNF-α in supernatants was variable. IL-12, IL-2, IL-4, IL-5 and IL-13 were not detected at significant concentrations. Galactocerebrosidase, transforming growth factor-β and IL-10 transcripts were specifically enriched in DUOC-01 relative to CB cells. PB MNCs proliferated and released cytokines in response to DUOC-01. DUOC-01 did not proliferate in response to mismatched MNC. CONCLUSIONS DUOC-01 has potential as an adjunctive cell therapy to myeloablative CB transplant for treatment of inherited metabolic diseases.
Collapse
Affiliation(s)
- Joanne Kurtzberg
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Susan Buntz
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Tracy Gentry
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Pamela Noeldner
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - April Ozamiz
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Benjamin Rusche
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert W Storms
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Amy Wollish
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - David A Wenger
- Lysosomal Diseases Testing Laboratory, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Andrew E Balber
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
526
|
Sun J, Furio L, Mecheri R, van der Does AM, Lundeberg E, Saveanu L, Chen Y, van Endert P, Agerberth B, Diana J. Pancreatic β-Cells Limit Autoimmune Diabetes via an Immunoregulatory Antimicrobial Peptide Expressed under the Influence of the Gut Microbiota. Immunity 2015; 43:304-17. [PMID: 26253786 DOI: 10.1016/j.immuni.2015.07.013] [Citation(s) in RCA: 212] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 05/15/2015] [Accepted: 05/22/2015] [Indexed: 12/14/2022]
Abstract
Antimicrobial peptides (AMPs) expressed by epithelial and immune cells are largely described for the defense against invading microorganisms. Recently, their immunomodulatory functions have been highlighted in various contexts. However how AMPs expressed by non-immune cells might influence autoimmune responses in peripheral tissues, such as the pancreas, is unknown. Here, we found that insulin-secreting β-cells produced the cathelicidin related antimicrobial peptide (CRAMP) and that this production was defective in non-obese diabetic (NOD) mice. CRAMP administrated to prediabetic NOD mice induced regulatory immune cells in the pancreatic islets, dampening the incidence of autoimmune diabetes. Additional investigation revealed that the production of CRAMP by β-cells was controlled by short-chain fatty acids produced by the gut microbiota. Accordingly, gut microbiota manipulations in NOD mice modulated CRAMP production and inflammation in the pancreatic islets, revealing that the gut microbiota directly shape the pancreatic immune environment and autoimmune diabetes development.
Collapse
Affiliation(s)
- Jia Sun
- State Key Laboratory of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition and School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue Wuxi, 214122 Jiangsu, P.R. China
| | - Laetitia Furio
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 1163, Institut Imagine, 24 Boulevard du Montparnasse, 75015 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 12 Rue de l'École de Médecine, 75006 Paris, France
| | - Ramine Mecheri
- Université Paris Descartes, Sorbonne Paris Cité, 12 Rue de l'École de Médecine, 75006 Paris, France
| | - Anne M van der Does
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Erik Lundeberg
- Department of Physiology and Pharmacology, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Loredana Saveanu
- Université Paris Descartes, Sorbonne Paris Cité, 12 Rue de l'École de Médecine, 75006 Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 1151, Institut Necker-Enfants Malades (INEM), Centre National de la Recherche Scientifique, Unité 8253, 149 rue de Sèvres, 75015 Paris, France
| | - Yongquan Chen
- State Key Laboratory of Food Science and Technology, Synergetic Innovation Center of Food Safety and Nutrition and School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue Wuxi, 214122 Jiangsu, P.R. China
| | - Peter van Endert
- Université Paris Descartes, Sorbonne Paris Cité, 12 Rue de l'École de Médecine, 75006 Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 1151, Institut Necker-Enfants Malades (INEM), Centre National de la Recherche Scientifique, Unité 8253, 149 rue de Sèvres, 75015 Paris, France
| | - Birgitta Agerberth
- Medical Microbial Pathogenesis Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet Karolinska University Hospital, SE-141 86 Stockholm, Sweden
| | - Julien Diana
- Université Paris Descartes, Sorbonne Paris Cité, 12 Rue de l'École de Médecine, 75006 Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 1151, Institut Necker-Enfants Malades (INEM), Centre National de la Recherche Scientifique, Unité 8253, 149 rue de Sèvres, 75015 Paris, France.
| |
Collapse
|
527
|
Liu SL, Peng BJ, Zhong YL, Liu YL, Song Z, Wang Z. Effect of 5-caffeoylquinic acid on the NF-κB signaling pathway, peroxisome proliferator-activated receptor gamma 2, and macrophage infiltration in high-fat diet-fed Sprague-Dawley rat adipose tissue. Food Funct 2015; 6:2779-86. [PMID: 26158223 DOI: 10.1039/c5fo00626k] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obesity, considered as a consequence of overnutrition, sustains a low-degree inflammatory state and results in insulin-resistance and type 2 diabetes. Here, we investigated the anti-inflammatory effects of 5-caffeoylquinic acid (5-CQA) in high-fat diet-induced obese rats. Serum interleukin (IL)-6, monocyte chemotactic protein 1 (MCP-1), tumor necrosis factor-alpha (TNF-α), total cholesterol (TC), triglyceride (TG), and free fatty acid (FFA) levels were determined. Expression of genes related to TG metabolism, macrophage biomarkers, and inflammation was assessed by real-time PCR. Protein expression of NF-κB, PPARγ2, and phosphorylated IκBα was evaluated by western blotting, and the histology of adipose tissue was examined. Supplementation of the rat diet with 5-CQA reduced obesity development, macrophage infiltration, and steatosis. Additionally, 5-CQA decreased the expression of NF-κB and downstream inflammatory cytokines, but increased the expression of PPARγ2, in a dose-dependent manner. Thus, 5-CQA improved obesity and obesity-related metabolic disturbances via PPARγ2 and the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Si-Le Liu
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, China.
| | | | | | | | | | | |
Collapse
|
528
|
Santoro A, Ferrante MC, Di Guida F, Pirozzi C, Lama A, Simeoli R, Clausi MT, Monnolo A, Mollica MP, Mattace Raso G, Meli R. Polychlorinated Biphenyls (PCB 101, 153, and 180) Impair Murine Macrophage Responsiveness to Lipopolysaccharide: Involvement of NF-κB Pathway. Toxicol Sci 2015; 147:255-69. [PMID: 26141388 PMCID: PMC7107302 DOI: 10.1093/toxsci/kfv127] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Non-dioxin-like (NDL) polychlorinated biphenyls (PCBs) are persistent organic pollutants, associated with a range of adverse health effects, including interference with the immune system. In this study, we investigate the capability of NDL-PCBs 101, 153, and 180, 3 of the 6 NDL-PCBs defined as indicators, to impair the immune response in lipopolysaccharide (LPS)-activated J774A.1 and primary murine macrophages. Our results clearly demonstrate that the exposure of J774A.1 and primary macrophages to NDL-PCB 153 or 180 or all NDL-PCBs mixtures causes a significant reduction in LPS-induced cytokine/chemokine synthesis, such as tumor necrosis factor-α and interleukin-6, together with monocyte chemoattractant protein-1, involved in cell recruitment. Moreover, PCBs were found to suppress LPS-stimulated NO production, and to reduce cyclooxygenase-2 and inducible nitric oxide synthase expression in J774A.1 and primary macrophages. At mechanistic level, PCBs significantly counteract the LPS-driven toll-like receptor (TLR) 4 and CD14 upregulation, therefore inhibiting downstream nuclear factor-κB (NF-κB) activation in J774A.1. Furthermore, PCBs determine a significant loss of macrophage endocytic capacity, a prerequisite for efficient antigen presentation. Taken together, these data indicate that NDL-PCBs reduce macrophage responsiveness, particularly when they are combined at concentrations per se inactive, impairing the capability to orchestrate a proper immune response to an infectious stimulus, disrupting TLR4/NF-κB pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Maria Pina Mollica
- Department of Biology, University of Naples Federico II, 80131 Naples, Italy
| | | | | |
Collapse
|
529
|
Liu Q, Rehman H, Krishnasamy Y, Schnellmann RG, Lemasters JJ, Zhong Z. Improvement of liver injury and survival by JNK2 and iNOS deficiency in liver transplants from cardiac death mice. J Hepatol 2015; 63:68-74. [PMID: 25703084 PMCID: PMC4475508 DOI: 10.1016/j.jhep.2015.02.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND & AIMS Inclusion of liver grafts from cardiac death donors (CDD) would increase the availability of donor livers but is hampered by a higher risk of primary non-function. Here, we seek to determine mechanisms that contribute to primary non-function of liver grafts from CDD with the goal to develop strategies for improved function and outcome, focusing on c-Jun-N-terminal kinase (JNK) activation and mitochondrial depolarization, two known mediators of graft failure. METHODS Livers explanted from wild-type, inducible nitric oxide synthase knockout (iNOS(-/-)), JNK1(-/-) or JNK2(-/-) mice after 45-min aorta clamping were implanted into wild-type recipients. Mitochondrial depolarization was detected by intravital confocal microscopy in living recipients. RESULTS After transplantation of wild-type CDD livers, graft iNOS expression and 3-nitrotyrosine adducts increased, but hepatic endothelial NOS expression was unchanged. Graft injury and dysfunction were substantially higher in CDD grafts than in non-CDD grafts. iNOS deficiency and inhibition attenuated injury and improved function and survival of CDD grafts. JNK1/2 and apoptosis signal-regulating kinase-1 activation increased markedly in wild-type CDD grafts, which was blunted by iNOS deficiency. JNK inhibition and JNK2 deficiency, but not JNK1 deficiency, decreased injury and improved function and survival of CDD grafts. Mitochondrial depolarization and binding of phospho-JNK2 to Sab, a mitochondrial protein linked to the mitochondrial permeability transition, were higher in CDD than in non-CDD grafts. iNOS deficiency, JNK inhibition and JNK2 deficiency all decreased mitochondrial depolarization and blunted ATP depletion in CDD grafts. JNK inhibition and deficiency did not decrease 3-nitrotyrosine adducts in CDD grafts. CONCLUSION The iNOS-JNK2-Sab pathway promotes CDD graft failure via increased mitochondrial depolarization, and is an attractive target to improve liver function and survival in CDD liver transplantation recipients.
Collapse
Affiliation(s)
- Qinlong Liu
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States; The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Hasibur Rehman
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Yasodha Krishnasamy
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Rick G Schnellmann
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States; Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States
| | - John J Lemasters
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Zhi Zhong
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC 29425, United States.
| |
Collapse
|
530
|
Grayfer L, Robert J. Distinct functional roles of amphibian (Xenopus laevis) colony-stimulating factor-1- and interleukin-34-derived macrophages. J Leukoc Biol 2015; 98:641-9. [PMID: 26136505 DOI: 10.1189/jlb.4ab0315-117rr] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/26/2015] [Indexed: 12/17/2022] Open
Abstract
Although Mϕ represent the most primordial immune cell subsets, the mechanisms governing their functional heterogeneity remain poorly defined. However, it is well established that the CSF-1 cytokine contributes to monopoiesis and to this heterogeneity, whereas the unrelated IL-34 also binds the CSF-1R toward poorly understood immunologic roles. To delineate the molecular and evolutionary basis behind vertebrate Mϕ functional heterogeneity, we performed comprehensive transcriptional and functional studies of amphibian (Xenopus laevis) BM (in vitro) and PER (in vivo) Mϕ derived by rXlCSF-1 and rXlIL-34. Our findings indicate that these amphibian cytokines promote morphologically and functionally distinct Mϕ populations. Mϕ induced by rXlCSF-1 possess more robust iNOS gene expression, are substantially more phagocytic, display greater NO responses, and exhibit enhanced bactericidal capacities. By contrast, rXlIL-34-derived Mϕ express greater levels of Arg-1 and NADPH oxidase components and possess greater respiratory burst responses. Most notably, whereas CSF-1 Mϕ are highly susceptible to the emerging FV3 ranavirus, rXlIL-34 Mϕ exhibit potent antiviral activity against this pathogen, which is dependent on reactive oxygen production. This work marks an advance in our understanding of the possible mechanisms governing vertebrate Mϕ functional heterogeneity.
Collapse
Affiliation(s)
- Leon Grayfer
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Jacques Robert
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
531
|
LAPUC IZABELA, BOLKUN LUKASZ, ELJASZEWICZ ANDRZEJ, RUSAK MALGORZATA, LUKSZA EWA, SINGH PAULINA, MIKLASZ PAULA, PISZCZ JAROSLAW, PTASZYNSKA-KOPCZYNSKA KATARZYNA, JASIEWICZ MALGORZATA, KAMINSKI KAROL, DABROWSKA MILENA, BODZENTA-LUKASZYK ANNA, KLOCZKO JANUSZ, MONIUSZKO MARCIN. Circulating classical CD14++CD16− monocytes predict shorter time to initial treatment in chronic lymphocytic leukemia patients: Differential effects of immune chemotherapy on monocyte-related membrane and soluble forms of CD163. Oncol Rep 2015; 34:1269-78. [PMID: 26135617 DOI: 10.3892/or.2015.4088] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/29/2015] [Indexed: 11/06/2022] Open
|
532
|
Franco R, Fernández-Suárez D. Alternatively activated microglia and macrophages in the central nervous system. Prog Neurobiol 2015; 131:65-86. [PMID: 26067058 DOI: 10.1016/j.pneurobio.2015.05.003] [Citation(s) in RCA: 495] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 05/22/2015] [Accepted: 05/30/2015] [Indexed: 12/20/2022]
Abstract
Macrophages are important players in the fight against viral, bacterial, fungal and parasitic infections. From a resting state they may undertake two activation pathways, the classical known as M1, or the alternative known as M2. M1 markers are mostly mediators of pro-inflammatory responses whereas M2 markers emerge for resolution and cleanup. Microglia exerts in the central nervous system (CNS) a function similar to that of macrophages in the periphery. Microglia activation and proliferation occurs in almost any single pathology affecting the CNS. Often microglia activation has been considered detrimental and drugs able to stop microglia activation were considered for the treatment of a variety of diseases. Cumulative evidence shows that microglia may undergo the alternative activation pathway, express M2-type markers and contribute to neuroprotection. This review focuses on details about the role of M2 microglia and in the approaches available for its identification. Approaches to drive the M2 phenotype and data on its potential in CNS diseases are also reviewed.
Collapse
Affiliation(s)
- Rafael Franco
- Molecular Neurobiology Laboratory, Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain; Centro Investigación Biomédica en Red: Enfermedades Neurodegenerativas (CIBERNED), Spain.
| | - Diana Fernández-Suárez
- Division of Molecular Neurobiology, Department of Neuroscience, Karolinska Institute, 17177 Stockholm, Sweden.
| |
Collapse
|
533
|
Kurtzberg J, Buntz S, Gentry T, Noeldner P, Ozamiz A, Rusche B, Storms RW, Wollish A, Wenger DA, Balber AE. Preclinical characterization of DUOC-01, a cell therapy product derived from banked umbilical cord blood for use as an adjuvant to umbilical cord blood transplantation for treatment of inherited metabolic diseases. Cytotherapy 2015; 17:803-815. [PMID: 25770677 PMCID: PMC4843803 DOI: 10.1016/j.jcyt.2015.02.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 02/09/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND AIMS Cord blood (CB) transplantation slows neurodegeneration during certain inherited metabolic diseases. However, the number of donor cells in the brain of patients does not appear to be sufficient to provide benefit until several months after transplant. We developed the cell product DUOC-01 to provide therapeutic effects in the early post-transplant period. METHODS DUOC-01 cultures initiated from banked CB units were characterized by use of time-lapse photomicroscopy during the 21-day manufacturing process. Antigen expression was measured by means of flow cytometry and immunocytochemistry; transcripts for cytokines and enzymes by quantitative real-time polymerase chain reaction; activities of lysosomal enzymes by direct biochemical analysis; alloreactivity of DUOC-01 and of peripheral blood (PB) mononuclear cells (MNC) to DUOC-01 by mixed lymphocyte culture methods; and cytokine secretion by Bioplex assays. RESULTS DUOC-01 cultures contained highly active, attached, motile, slowly proliferating cells that expressed common (cluster of differentiation [CD]11b, CD14 and Iba1), M1 type (CD16, inducible nitric oxide synthase), and M2-type (CD163, CD206) macrophage or microglia markers. Activities of 11 disease-relevant lysosomal enzymes in DUOC-01 products were similar to those of normal PB cells. All DUOC-01 products secreted interleukin (IL)-6 and IL-10. Accumulation of transforming growth factor-β, IL-1β, interferon-γ and TNF-α in supernatants was variable. IL-12, IL-2, IL-4, IL-5 and IL-13 were not detected at significant concentrations. Galactocerebrosidase, transforming growth factor-β and IL-10 transcripts were specifically enriched in DUOC-01 relative to CB cells. PB MNCs proliferated and released cytokines in response to DUOC-01. DUOC-01 did not proliferate in response to mismatched MNC. CONCLUSIONS DUOC-01 has potential as an adjunctive cell therapy to myeloablative CB transplant for treatment of inherited metabolic diseases.
Collapse
Affiliation(s)
- Joanne Kurtzberg
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Susan Buntz
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Tracy Gentry
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Pamela Noeldner
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - April Ozamiz
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Benjamin Rusche
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert W Storms
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Amy Wollish
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - David A Wenger
- Lysosomal Diseases Testing Laboratory, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Andrew E Balber
- Robertson Clinical and Translational Cell Therapy Program, Duke Translational Medicine Institute, Duke University Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
534
|
Advanced Glycation End Products Enhance Macrophages Polarization into M1 Phenotype through Activating RAGE/NF-κB Pathway. BIOMED RESEARCH INTERNATIONAL 2015; 2015:732450. [PMID: 26114112 PMCID: PMC4465680 DOI: 10.1155/2015/732450] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 05/12/2015] [Indexed: 01/01/2023]
Abstract
Atherosclerotic lesions are accelerated in patients with diabetes. M1 (classically activated in contrast to M2 alternatively activated) macrophages play key roles in the progression of atherosclerosis. Since advanced glycation end products (AGEs) are major pathogenic factors and active inflammation inducers in diabetes mellitus, this study assessed the effects of AGEs on macrophage polarization. The present study showed that AGEs significantly promoted macrophages to express IL-6 and TNF-α. M1 macrophage markers such as iNOS and surface markers including CD11c and CD86 were significantly upregulated while M2 macrophage markers such as Arg1 and CD206 remained unchanged after AGEs stimulation. AGEs significantly increased RAGE expression in macrophages and activated NF-κB pathway, and the aforementioned effects were partly abolished by administration of anti-RAGE antibody or NF-κB inhibitor PDTC. In conclusion, our results suggest that AGEs enhance macrophage differentiation into proinflammatory M1 phenotype at least partly via RAGE/NF-κB pathway activation.
Collapse
|
535
|
Saha B, Bala S, Hosseini N, Kodys K, Szabo G. Krüppel-like factor 4 is a transcriptional regulator of M1/M2 macrophage polarization in alcoholic liver disease. J Leukoc Biol 2015; 97:963-973. [PMID: 25740962 PMCID: PMC6608000 DOI: 10.1189/jlb.4a1014-485r] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 01/29/2015] [Accepted: 02/11/2015] [Indexed: 12/12/2022] Open
Abstract
Macrophages play an important role in inflammation and liver injury. In ALD, activated macrophages, including M1 (proinflammatory) and M2 (anti-inflammatory) macrophages, are present in the liver. As KLF4 has been described as a regulator of macrophage polarization, we investigated its role in ALD. Chronic alcohol feeding in C57Bl/6 mice led to increased expression of M1 (TNF-α, MCP1, and IL-1β) and M2 (Arg1, Mrc1, and IL-10) genes and the frequency of CD206+CD163+ M2 macrophages in the liver. KLF4 mRNA and protein levels were increased in the livers of EtFed compared with PF mice. In macrophages, in vivo and in vitro, EtOH increased KLF4 levels, transcriptional activity, and expression of M1 and M2 genes. KLF4 knockdown and overexpression experiments demonstrated alcohol-dependent and -independent functions of KLF4 in regulating M1 and M2 markers. KLF4 siRNA treatment, alone and in synergy with alcohol, increased the levels of M1 markers. In contrast, KLF4 overexpression increased the levels of M2 and decreased M1 markers, and this was enhanced further by alcohol. KLF4 was regulated by alcohol and its metabolites. KLF4 mRNA and activity were increased in the presence of 4-MP, an inhibitor of ADH, and CYP2E1. However, inhibition of acetaldehyde breakdown attenuated KLF4 induction and promoted M1 polarization. We conclude that KLF4 regulates M1 and M2 markers in ALD. EtOH promotes KLF4 and M2 phenotype, whereas acetaldehyde attenuates KLF4 and promotes M1 macrophage, which may explain the increased presence of M1 and M2 macrophage populations in ALD.
Collapse
Affiliation(s)
- Banishree Saha
- University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Shashi Bala
- University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Nooshin Hosseini
- University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Karen Kodys
- University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Gyongyi Szabo
- University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
536
|
Kapoor N, Niu J, Saad Y, Kumar S, Sirakova T, Becerra E, Li X, Kolattukudy PE. Transcription factors STAT6 and KLF4 implement macrophage polarization via the dual catalytic powers of MCPIP. THE JOURNAL OF IMMUNOLOGY 2015; 194:6011-23. [PMID: 25934862 DOI: 10.4049/jimmunol.1402797] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 04/02/2015] [Indexed: 01/06/2023]
Abstract
Macrophage polarization plays a critical role in tissue homeostasis, disease pathogenesis, and inflammation and its resolution. IL-4-induced macrophage polarization involves induction of STAT6 and Krüppel-like factor 4 (KLF4), which induce each other and promote M2 polarization. However, how these transcription factors implement M2 polarization is not understood. We report that in murine macrophages MCP-1-induced protein (MCPIP), induced by KLF4, inhibits M1 polarization by inhibiting NF-κB activation and implements M2 polarization using both its deubiquitinase and RNase activities that cause sequential induction of reactive oxygen species (ROS), endoplasmic reticulum (ER) stress, and autophagy required for M2 polarization. MCPIP also induces C/EBPβ and PPARγ, which promote M2 polarization. Macrophages from mice with myeloid-targeted overexpression of MCPIP show elevated expression of M2 markers and reduced response to LPS, whereas macrophages from mice with myeloid-specific deletion of MCPIP manifest elevated M1 polarization with enhanced phagocytic activity. Thus, both in vivo and in vitro experiments demonstrate that the transcription factors STAT6 and KLF4 implement IL-4-induced M2 polarization via the dual catalytic activities of MCPIP.
Collapse
Affiliation(s)
- Nidhi Kapoor
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816
| | - Jianli Niu
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816
| | - Yasser Saad
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816
| | - Sanjay Kumar
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816
| | - Tatiana Sirakova
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816
| | - Edilu Becerra
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816
| | - Xiaoman Li
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816
| | - Pappachan E Kolattukudy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32816
| |
Collapse
|
537
|
Transcriptional programming of human macrophages: on the way to systems immunology. J Mol Med (Berl) 2015; 93:589-97. [PMID: 25877862 DOI: 10.1007/s00109-015-1286-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 03/09/2015] [Accepted: 03/12/2015] [Indexed: 12/13/2022]
Abstract
Many of the major common diseases such as atherosclerosis, diabetes, obesity, numerous autoimmune diseases, as well as neurodegenerative diseases such as Alzheimer's disease and many cancer types are characterised by a chronic inflammatory component termed sterile inflammation. Myeloid cells, particularly macrophages, are an important cellular component of chronic inflammation in these diseases. For almost all of these disease conditions, previous reports suggested that macrophages can exert either so-called pro-inflammatory or anti-inflammatory functions, thereby either fighting or feeding the disease. This apparent dichotomy of reactions of macrophages led to a dichotomous definition of macrophage activation classified as macrophage polarisation. However, analysis of large transcriptomics data derived from human and murine macrophages show that macrophage functions are shaped in a very tissue- and signal-input specific manner, allowing these cells to develop extremely specific functional programmes. Integrating global views on macrophage activation on the transcriptome, the epigenome, the proteome or the metabolome will finally lead to a data-driven approach to understand macrophage biology in context of major diseases. We are indeed on the way to a systems immunology approach that integrates -omics data with mathematical and bioinformatical modelling as the pre-requisite to generate data-driven hypotheses. This approach opens completely new avenues for the development of tailored diagnostics and therapies targeting macrophages in sterile inflammations of the major common diseases. I will also discuss some of the next developments that will be necessary to reach these important goals.
Collapse
|
538
|
Pivotal roles of GM-CSF in autoimmunity and inflammation. Mediators Inflamm 2015; 2015:568543. [PMID: 25838639 PMCID: PMC4370199 DOI: 10.1155/2015/568543] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/23/2015] [Indexed: 12/14/2022] Open
Abstract
Granulocyte macrophage-colony stimulating factor (GM-CSF) is a hematopoietic growth factor, which stimulates the proliferation of granulocytes and macrophages from bone marrow precursor cells. In autoimmune and inflammatory diseases, Th17 cells have been considered as strong inducers of tissue inflammation. However, recent evidence indicates that GM-CSF has prominent proinflammatory functions and that this growth factor (not IL-17) is critical for the pathogenicity of CD4+ T cells. Therefore, the mechanism of GM-CSF-producing CD4+ T cell differentiation and the role of GM-CSF in the development of autoimmune and inflammatory diseases are gaining increasing attention. This review summarizes the latest knowledge of GM-CSF and its relationship with autoimmune and inflammatory diseases. The potential therapies targeting GM-CSF as well as their possible side effects have also been addressed in this review.
Collapse
|
539
|
Z-100, extracted from Mycobacterium tuberculosis strain Aoyama B, promotes TNF-α production via nucleotide-binding oligomerization domain containing 2 (Nod2)-dependent NF-κB activation in RAW264.7 cells. Mol Immunol 2015; 64:218-27. [DOI: 10.1016/j.molimm.2014.11.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/20/2014] [Accepted: 11/25/2014] [Indexed: 01/07/2023]
|
540
|
Yue S, Mu W, Erb U, Zöller M. The tetraspanins CD151 and Tspan8 are essential exosome components for the crosstalk between cancer initiating cells and their surrounding. Oncotarget 2015; 6:2366-84. [PMID: 25544774 PMCID: PMC4385857 DOI: 10.18632/oncotarget.2958] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/09/2014] [Indexed: 01/01/2023] Open
Abstract
Tspan8 and CD151 are metastasis-promoting tetraspanins and a knockdown (kd) of Tspan8 or CD151 and most pronounced of both tetraspanins affects the metastatic potential of the rat pancreatic adenocarcinoma line ASML. Approaching to elaborate the underlying mechanism, we compared ASMLwt, -CD151kd and/or Tspan8kd clones. We focused on tumor exosomes, as exosomes play a major role in tumor progression and tetraspanins are suggested to be engaged in exosome targeting. ASML-CD151/Tspan8kd cells poorly metastasize, but regain metastatic capacity, when rats are pretreated with ASMLwt, but not ASML-CD151kd and/or -Tspan8kd exosomes. Both exosomal CD151 and Tspan8 contribute to host matrix remodelling due to exosomal tetraspanin-integrin and tetraspanin-protease associations. ASMLwt exosomes also support stroma cell activation with upregulation of cytokines, cytokine receptors and proteases and promote inflammatory cytokine expression in hematopoietic cells. Finally, CD151-/Tspan8-competent exosomes support EMT gene expression in poorly-metastatic ASML-CD151/Tspan8kd cells. These effects are not seen or are weakened using ASML-CD151kd or -Tspan8kd exosomes, which is at least partly due to reduced binding/uptake of CD151- and/or Tspan8-deficient exosomes. Thus, CD151- and Tspan8-competent tumor exosomes support matrix degradation, reprogram stroma and hematopoietic cells and drive non-metastatic ASML-CD151/Tspan8kd cells towards a motile phenotype.
Collapse
Affiliation(s)
- Shijing Yue
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Wei Mu
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Ulrike Erb
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| | - Margot Zöller
- Department of Tumor Cell Biology, University Hospital of Surgery, Heidelberg, Germany
| |
Collapse
|
541
|
Wermuth PJ, Jimenez SA. The significance of macrophage polarization subtypes for animal models of tissue fibrosis and human fibrotic diseases. Clin Transl Med 2015; 4:2. [PMID: 25852818 PMCID: PMC4384891 DOI: 10.1186/s40169-015-0047-4] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 01/20/2015] [Indexed: 12/15/2022] Open
Abstract
The systemic and organ-specific human fibrotic disorders collectively represent one of the most serious health problems world-wide causing a large proportion of the total world population mortality. The molecular pathways involved in their pathogenesis are complex and despite intensive investigations have not been fully elucidated. Whereas chronic inflammatory cell infiltration is universally present in fibrotic lesions, the central role of monocytes and macrophages as regulators of inflammation and fibrosis has only recently become apparent. However, the precise mechanisms involved in the contribution of monocytes/macrophages to the initiation, establishment, or progression of the fibrotic process remain largely unknown. Several monocyte and macrophage subpopulations have been identified, with certain phenotypes promoting inflammation whereas others display profibrotic effects. Given the unmet need for effective treatments for fibroproliferative diseases and the crucial regulatory role of monocyte/macrophage subpopulations in fibrogenesis, the development of therapeutic strategies that target specific monocyte/macrophage subpopulations has become increasingly attractive. We will provide here an overview of the current understanding of the role of monocyte/macrophage phenotype subpopulations in animal models of tissue fibrosis and in various systemic and organ-specific human fibrotic diseases. Furthermore, we will discuss recent approaches to the design of effective anti-fibrotic therapeutic interventions by targeting the phenotypic differences identified between the various monocyte and macrophage subpopulations.
Collapse
Affiliation(s)
- Peter J Wermuth
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Bluemle Life Science Building Suite 509, 233 South 10th Street, Philadelphia, PA 19107-5541 USA
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Bluemle Life Science Building Suite 509, 233 South 10th Street, Philadelphia, PA 19107-5541 USA
| |
Collapse
|
542
|
Schönheit J, Leutz A, Rosenbauer F. Chromatin Dynamics during Differentiation of Myeloid Cells. J Mol Biol 2015; 427:670-87. [DOI: 10.1016/j.jmb.2014.08.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/05/2014] [Accepted: 08/20/2014] [Indexed: 12/23/2022]
|
543
|
Cassano JM, Schnabel LV, Betancourt AM, Antczak DF, Fortier LA. Mesenchymal Stem Cell Therapy: Clinical Progress and Opportunities for Advancement. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0064-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
544
|
Rojas J, Salazar J, Martínez MS, Palmar J, Bautista J, Chávez-Castillo M, Gómez A, Bermúdez V. Macrophage Heterogeneity and Plasticity: Impact of Macrophage Biomarkers on Atherosclerosis. SCIENTIFICA 2015; 2015:851252. [PMID: 26491604 PMCID: PMC4600540 DOI: 10.1155/2015/851252] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/09/2015] [Indexed: 05/15/2023]
Abstract
Cardiovascular disease (CVD) is a global epidemic, currently representing the worldwide leading cause of morbidity and mortality. Atherosclerosis is the fundamental pathophysiologic component of CVD, where the immune system plays an essential role. Monocytes and macrophages are key mediators in this aspect: due to their heterogeneity and plasticity, these cells may act as either pro- or anti-inflammatory mediators. Indeed, monocytes may develop heterogeneous functional phenotypes depending on the predominating pro- or anti-inflammatory microenvironment within the lesion, resulting in classic, intermediate, and non-classic monocytes, each with strikingly differing features. Similarly, macrophages may also adopt heterogeneous profiles being mainly M1 and M2, the former showing a proinflammatory profile while the latter demonstrates anti-inflammatory traits; they are further subdivided in several subtypes with more specialized functions. Furthermore, macrophages may display plasticity by dynamically shifting between phenotypes in response to specific signals. Each of these distinct cell profiles is associated with diverse biomarkers which may be exploited for therapeutic intervention, including IL-10, IL-13, PPAR-γ, LXR, NLRP3 inflammasomes, and microRNAs. Direct modulation of the molecular pathways concerning these potential macrophage-related targets represents a promising field for new therapeutic alternatives in atherosclerosis and CVD.
Collapse
Affiliation(s)
- Joselyn Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
- Endocrinology Department, Maracaibo University Hospital, Maracaibo 4004, Venezuela
- *Joselyn Rojas:
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - María Sofía Martínez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Jim Palmar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Jordan Bautista
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Mervin Chávez-Castillo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Alexis Gómez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| | - Valmore Bermúdez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo 4004, Venezuela
| |
Collapse
|
545
|
Abstract
Macrophage involvement in viral infections and antiviral states is common. However, this involvement has not been well-studied in the paradigm of macrophage polarization, which typically has been categorized by the dichotomy of classical (M1) and alternative (M2) statuses. Recent studies have revealed the complexity of macrophage polarization in response to various cellular mediators and exogenous stimuli by adopting a multipolar view to revisit the differential process of macrophages, especially those re-polarized during viral infections. Here, through examination of viral infections targeting macrophages/monocytic cells, we focus on the direct involvement of macrophage polarization during viral infections. Type I and type III interferons (IFNs) are critical in regulation of viral pathogenesis and host antiviral infection; thus, we propose to incorporate IFN-mediated antiviral states into the framework of macrophage polarization. This view is supported by the multifunctional properties of type I IFNs, which potentially elicit and regulate both M1- and M2-polarization in addition to inducing the antiviral state, and by the discoveries of viral mechanisms to adapt and modulate macrophage polarization. Indeed, several recent studies have demonstrated effective prevention of viral diseases through manipulation of macrophage immune statuses.
Collapse
Affiliation(s)
- Yongming Sang
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Laura C Miller
- Virus and Prion Diseases of Livestock Research Unit, National Animal Disease Center, USDA-ARS, 1920 Dayton Ave, Ames, IA 50010, USA
| | - Frank Blecha
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
546
|
Krist B, Florczyk U, Pietraszek-Gremplewicz K, Józkowicz A, Dulak J. The Role of miR-378a in Metabolism, Angiogenesis, and Muscle Biology. Int J Endocrinol 2015; 2015:281756. [PMID: 26839547 PMCID: PMC4709675 DOI: 10.1155/2015/281756] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/30/2015] [Indexed: 02/06/2023] Open
Abstract
MicroRNA-378a (miR-378a, previously known as miR-378) is one of the small noncoding RNA molecules able to regulate gene expression at posttranscriptional level. Its two mature strands, miR-378a-3p and miR-378a-5p, originate from the first intron of the peroxisome proliferator-activated receptor gamma, coactivator 1 beta (ppargc1b) gene encoding PGC-1β. Embedding in the sequence of this transcriptional regulator of oxidative energy metabolism implies involvement of miR-378a in metabolic pathways, mitochondrial energy homeostasis, and related biological processes such as muscle development, differentiation, and regeneration. On the other hand, modulating the expression of proangiogenic factors such as vascular endothelial growth factor, angiopoietin-1, or interleukin-8, influencing inflammatory reaction, and affecting tumor suppressors, such as SuFu and Fus-1, miR-378a is considered as a part of an angiogenic network in tumors. In the latter, miR-378a can evoke broader actions by enhancing cell survival, reducing apoptosis, and promoting cell migration and invasion. This review describes the current knowledge on miR-378a linking oxidative/lipid metabolism, muscle biology, and blood vessel formation.
Collapse
Affiliation(s)
- Bart Krist
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Urszula Florczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Katarzyna Pietraszek-Gremplewicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
- *Jozef Dulak:
| |
Collapse
|
547
|
Fricke S, Pfefferkorn C, Wolf D, Riemschneider S, Kohlschmidt J, Hilger N, Fueldner C, Knauer J, Sack U, Emmrich F, Lehmann J. Characterization of the murine myeloid precursor cell line MuMac-E8. PLoS One 2014; 9:e113743. [PMID: 25546418 PMCID: PMC4278753 DOI: 10.1371/journal.pone.0113743] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 10/30/2014] [Indexed: 12/15/2022] Open
Abstract
Starting point for the present work was the assumption that the cell line MuMac-E8 represents a murine cell population with stem cell properties. Preliminary studies already pointed to the expression of stem-cell associated markers and a self-regenerative potential of the cells. The cell line MuMac-E8 should be examined for their differential stage within stem cell hierarchy. MuMac-E8 cells were derived from a chimeric mouse model of arthritis. It could be shown that MuMac-E8 cells express mRNA of some genes associated with pluripotent stem cells (Nanog, Nucleostemin), of genes for hematopoietic markers (EPCR, Sca-1, CD11b, CD45), for the mesenchymal marker CD105 and of genes for the neural markers Pax-6 and Ezrin. In methylcellulose and May-Grünwald-Giemsa staining, hematopoietic colonies were obtained but the hematopoietic system of lethally irradiated mice could not be rescued. Osteogenic differentiation was not detectable. Thus, it became evident that MuMac-E8 represents not a stem cell line. However, MuMac-E8 cells expressed several myeloid surface markers (i.e. CD11b, F4/80, CD14, CD64), showed phagocytosis and is capable of producing nitric oxide. Thus, this cell line seems to be arrested an advanced stage of myeloid differentiation. Adherence data measured by impedance-based real-time cell analysis together with cell morphology data suggested that MuMac-E8 represents a new macrophage precursor cell line exhibiting weak adherence. This cell line is suitable as an in-vitro model for testing of macrophage functions. Moreover, it might be also useful for differentiation or reprogramming studies.
Collapse
Affiliation(s)
- Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
- Translational Centre for Regenerative Medicine, University of Leipzig, Leipzig, Germany
| | | | - Doris Wolf
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Department of Surgery, Research Laboratories, University of Leipzig, Leipzig, Germany
| | - Sina Riemschneider
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Janine Kohlschmidt
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Nadja Hilger
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Christiane Fueldner
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Jens Knauer
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
| | - Ulrich Sack
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
- Translational Centre for Regenerative Medicine, University of Leipzig, Leipzig, Germany
| | - Frank Emmrich
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
- Translational Centre for Regenerative Medicine, University of Leipzig, Leipzig, Germany
| | - Jörg Lehmann
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- * E-mail:
| |
Collapse
|
548
|
Zhu YP, Brown JR, Sag D, Zhang L, Suttles J. Adenosine 5'-monophosphate-activated protein kinase regulates IL-10-mediated anti-inflammatory signaling pathways in macrophages. THE JOURNAL OF IMMUNOLOGY 2014; 194:584-94. [PMID: 25512602 DOI: 10.4049/jimmunol.1401024] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AMP-activated protein kinase (AMPK) is a conserved serine/threonine kinase with a critical function in the regulation of metabolic pathways in eukaryotic cells. Recently, AMPK has been shown to play an additional role as a regulator of inflammatory activity in leukocytes. Treatment of macrophages with chemical AMPK activators, or forced expression of a constitutively active form of AMPK, results in polarization to an anti-inflammatory phenotype. In addition, we reported previously that stimulation of macrophages with anti-inflammatory cytokines such as IL-10, IL-4, and TGF-β results in rapid activation of AMPK, suggesting that AMPK contributes to the suppressive function of these cytokines. In this study, we investigated the role of AMPK in IL-10-induced gene expression and anti-inflammatory function. IL-10-stimulated wild-type macrophages displayed rapid activation of PI3K and its downstream targets Akt and mammalian target of rapamycin complex (mTORC1), an effect that was not seen in macrophages generated from AMPKα1-deficient mice. AMPK activation was not impacted by treatment with either the PI3K inhibitor LY294002 or the JAK inhibitor CP-690550, suggesting that IL-10-mediated activation of AMPK is independent of PI3K and JAK activity. IL-10 induced phosphorylation of both Tyr(705) and Ser(727) residues of STAT3 in an AMPKα1-dependent manner, and these phosphorylation events were blocked by inhibition of Ca(2+)/calmodulin-dependent protein kinase kinase β, an upstream activator of AMPK, and by the mTORC1 inhibitor rapamycin, respectively. The impaired STAT3 phosphorylation in response to IL-10 observed in AMPKα1-deficient macrophages was accompanied by reduced suppressor of cytokine signaling 3 expression and an inadequacy of IL-10 to suppress LPS-induced proinflammatory cytokine production. Overall, our data demonstrate that AMPKα1 is required for IL-10 activation of the PI3K/Akt/mTORC1 and STAT3-mediated anti-inflammatory pathways regulating macrophage functional polarization.
Collapse
Affiliation(s)
- Yanfang Peipei Zhu
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292
| | - Jonathan R Brown
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292
| | - Duygu Sag
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292
| | - Lihua Zhang
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292
| | - Jill Suttles
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40292
| |
Collapse
|
549
|
Huen SC, Huynh L, Marlier A, Lee Y, Moeckel GW, Cantley LG. GM-CSF Promotes Macrophage Alternative Activation after Renal Ischemia/Reperfusion Injury. J Am Soc Nephrol 2014; 26:1334-45. [PMID: 25388222 DOI: 10.1681/asn.2014060612] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 10/09/2014] [Indexed: 11/03/2022] Open
Abstract
After kidney ischemia/reperfusion (I/R) injury, monocytes home to the kidney and differentiate into activated macrophages. Whereas proinflammatory macrophages contribute to the initial kidney damage, an alternatively activated phenotype can promote normal renal repair. The microenvironment of the kidney during the repair phase mediates the transition of macrophage activation from a proinflammatory to a reparative phenotype. In this study, we show that macrophages isolated from murine kidneys during the tubular repair phase after I/R exhibit an alternative activation gene profile that differs from the canonical alternative activation induced by IL-4-stimulated STAT6 signaling. This unique activation profile can be reproduced in vitro by stimulation of bone marrow-derived macrophages with conditioned media from serum-starved mouse proximal tubule cells. Secreted tubular factors were found to activate macrophage STAT3 and STAT5 but not STAT6, leading to induction of the unique alternative activation pattern. Using STAT3-deficient bone marrow-derived macrophages and pharmacologic inhibition of STAT5, we found that tubular cell-mediated macrophage alternative activation is regulated by STAT5 activation. Both in vitro and after renal I/R, tubular cells expressed GM-CSF, a known STAT5 activator, and this pathway was required for in vitro alternative activation of macrophages by tubular cells. Furthermore, administration of a neutralizing antibody against GM-CSF after renal I/R attenuated kidney macrophage alternative activation and suppressed tubular proliferation. Taken together, these data show that tubular cells can instruct macrophage activation by secreting GM-CSF, leading to a unique macrophage reparative phenotype that supports tubular proliferation after sterile ischemic injury.
Collapse
Affiliation(s)
| | | | | | - Yashang Lee
- Section of Nephrology, Department of Medicine
| | - Gilbert W Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | | |
Collapse
|
550
|
KANG FUBIAO, WANG LING, LI DONG, ZHANG YINGE, SUN DIANXING. Hepatocellular carcinomas promote tumor-associated macrophage M2-polarization via increased B7-H3 expression. Oncol Rep 2014; 33:274-82. [DOI: 10.3892/or.2014.3587] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/08/2014] [Indexed: 11/06/2022] Open
|