401
|
Liu L, Hu F, Wang H, Wu X, Eltahan AS, Stanford S, Bottini N, Xiao H, Bottini M, Guo W, Liang XJ. Secreted Protein Acidic and Rich in Cysteine Mediated Biomimetic Delivery of Methotrexate by Albumin-Based Nanomedicines for Rheumatoid Arthritis Therapy. ACS NANO 2019; 13:5036-5048. [PMID: 30978282 DOI: 10.1021/acsnano.9b01710] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Rheumatoid arthritis (RA) is one of the most common chronic autoimmune diseases. Despite considerable advances in clinical treatment of RA, suboptimal response to therapy and treatment discontinuation are still unresolved challenges due to systemic toxicity. It is of crucial importance to actively target and deliver therapeutic agents to inflamed joints in order to promote in situ activity and decrease systemic toxicity. In this study, we found that SPARC (secreted protein acidic and rich in cysteine) was overexpressed in the synovial fluid and synovium of RA patients as well as mice with collagen-induced arthritis (CIA), which has been scarcely reported. Building upon the SPARC signature of RA joint microenvironment and the intrinsic high affinity of SPARC for albumin, we fabricated methotrexate-loaded human serum albumin nanomedicines (MTX@HSA NMs) and explored them as biomimetic drug delivery systems for RA therapy. Upon intravenous injection of chlorin e6-labeled MTX@HSA NMs into CIA mice, the fluorescence/magnetic resonance dual-modal imaging revealed higher accumulations and longer retention of MTX@HSA NMs in inflamed joints with respect to free MTX molecules. In vivo therapeutic evaluations suggested that the MTX@HSA NMs were able to attenuate the progression of RA with better efficacy and fewer side effects even at half dose of administrated MTX in comparison with free MTX. By unraveling the mechanism driving the efficient accumulation of MTX@HSA NMs in RA joints and showing their ability to improve the safety and therapeutic efficacy of MTX, our work sheds light on the development of innovative anti-RA nanomedicines with a strong potential for clinical translation.
Collapse
Affiliation(s)
- Lu Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , People's Republic of China
- University of Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
- Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Rome , 00133 , Italy
| | - Fanlei Hu
- Department of Rheumatology and Immunology , Peking University People's Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis , Beijing 100044 , People's Republic of China
| | - Hui Wang
- CAS Key Laboratory for Nanosystem and Hierarchy Fabrication, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology , Chinese Academy of Sciences , Beijing 100190 , People's Republic of China
| | - Xiaoli Wu
- School of Life Sciences , Tianjin University , Tianjin 300072 , People's Republic of China
| | - Ahmed Shaker Eltahan
- Translational Medicine Center, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital , Guangzhou Medical University , Guangzhou 510260 , People's Republic of China
| | - Stephanie Stanford
- Altman Clinical & Translational Research Institute , University of California San Diego , La Jolla , California 92037 , United States
| | - Nunzio Bottini
- Altman Clinical & Translational Research Institute , University of California San Diego , La Jolla , California 92037 , United States
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry , Chinese Academy of Sciences , Beijing 100190 , People's Republic of China
| | - Massimo Bottini
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , People's Republic of China
- Department of Experimental Medicine and Surgery , University of Rome Tor Vergata , Rome , 00133 , Italy
| | - Weisheng Guo
- Translational Medicine Center, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital , Guangzhou Medical University , Guangzhou 510260 , People's Republic of China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , Beijing 100190 , People's Republic of China
- University of Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| |
Collapse
|
402
|
Kuo D, Ding J, Cohn IS, Zhang F, Wei K, Rao DA, Rozo C, Sokhi UK, Shanaj S, Oliver DJ, Echeverria AP, DiCarlo EF, Brenner MB, Bykerk VP, Goodman SM, Raychaudhuri S, Rätsch G, Ivashkiv LB, Donlin LT. HBEGF + macrophages in rheumatoid arthritis induce fibroblast invasiveness. Sci Transl Med 2019; 11:eaau8587. [PMID: 31068444 PMCID: PMC6726376 DOI: 10.1126/scitranslmed.aau8587] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 12/22/2018] [Accepted: 04/19/2019] [Indexed: 12/12/2022]
Abstract
Macrophages tailor their function according to the signals found in tissue microenvironments, assuming a wide spectrum of phenotypes. A detailed understanding of macrophage phenotypes in human tissues is limited. Using single-cell RNA sequencing, we defined distinct macrophage subsets in the joints of patients with the autoimmune disease rheumatoid arthritis (RA), which affects ~1% of the population. The subset we refer to as HBEGF+ inflammatory macrophages is enriched in RA tissues and is shaped by resident fibroblasts and the cytokine tumor necrosis factor (TNF). These macrophages promoted fibroblast invasiveness in an epidermal growth factor receptor-dependent manner, indicating that intercellular cross-talk in this inflamed setting reshapes both cell types and contributes to fibroblast-mediated joint destruction. In an ex vivo synovial tissue assay, most medications used to treat RA patients targeted HBEGF+ inflammatory macrophages; however, in some cases, medication redirected them into a state that is not expected to resolve inflammation. These data highlight how advances in our understanding of chronically inflamed human tissues and the effects of medications therein can be achieved by studies on local macrophage phenotypes and intercellular interactions.
Collapse
Affiliation(s)
- David Kuo
- Graduate Program in Physiology, Biophysics and Systems Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Computational Biology Program, Sloan Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Jennifer Ding
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Ian S Cohn
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Fan Zhang
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA 02115, USA
- Division of Rheumatology and Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kevin Wei
- Division of Rheumatology, Immunology, Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Deepak A Rao
- Division of Rheumatology, Immunology, Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Cristina Rozo
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Upneet K Sokhi
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Sara Shanaj
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - David J Oliver
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Adriana P Echeverria
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Edward F DiCarlo
- Department of Pathology and Laboratory Medicine, Hospital for Special Surgery, New York, NY 10021, USA
| | - Michael B Brenner
- Division of Rheumatology, Immunology, Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Vivian P Bykerk
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
- Weill Cornell Medical College, New York, NY 10021, USA
| | - Susan M Goodman
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
- Weill Cornell Medical College, New York, NY 10021, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA 02115, USA
- Division of Rheumatology and Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, UK
| | - Gunnar Rätsch
- Department of Computer Science, Universitätstrasse 6, ETH Zürich, 8092 Zürich, Switzerland
- Computational Biology Program, Sloan Kettering Institute, 1275 York Avenue, New York, NY 10065, USA
| | - Lionel B Ivashkiv
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
- Weill Cornell Medical College, New York, NY 10021, USA
- Weill Cornell Graduate School of Medical Sciences, New York, NY 10021, USA
| | - Laura T Donlin
- Arthritis and Tissue Degeneration Program and the David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA.
- Weill Cornell Medical College, New York, NY 10021, USA
| |
Collapse
|
403
|
Mantovani A, Dinarello CA, Molgora M, Garlanda C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019; 50:778-795. [PMID: 30995499 PMCID: PMC7174020 DOI: 10.1016/j.immuni.2019.03.012] [Citation(s) in RCA: 692] [Impact Index Per Article: 115.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023]
Abstract
Forty years after its naming, interleukin-1 (IL-1) is experiencing a renaissance brought on by the growing understanding of its context-dependent roles and advances in the clinic. Recent studies have identified important roles for members of the IL-1 family-IL-18, IL-33, IL-36, IL-37, and IL-38-in inflammation and immunity. Here, we review the complex functions of IL-1 family members in the orchestration of innate and adaptive immune responses and their diversity and plasticity. We discuss the varied roles of IL-1 family members in immune homeostasis and their contribution to pathologies, including autoimmunity and auto-inflammation, dysmetabolism, cardiovascular disorders, and cancer. The trans-disease therapeutic activity of anti-IL-1 strategies argues for immunity and inflammation as a metanarrative of modern medicine.
Collapse
Affiliation(s)
- Alberto Mantovani
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy; Humanitas University, via Rita Levi Montalcini, 20090 Pieve Emanuele Milan, Italy; William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martina Molgora
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy; Humanitas University, via Rita Levi Montalcini, 20090 Pieve Emanuele Milan, Italy.
| |
Collapse
|
404
|
Shen Y, Song J, Wang Y, Chen Z, Zhang L, Yu J, Zhu D, Zhong M. M2 macrophages promote pulmonary endothelial cells regeneration in sepsis-induced acute lung injury. ANNALS OF TRANSLATIONAL MEDICINE 2019; 7:142. [PMID: 31157263 DOI: 10.21037/atm.2019.02.47] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Background Macrophages can polarize to M2 phenotype to decrease inflammation and encourage tissue repair. Nonetheless, its role in sepsis-induced acute lung injury and its effect on endothelial cells (ECs) regeneration remains unknown. The aim of the current study was to explore the impact of M2 macrophages on pulmonary ECs proliferation in sepsis-induced acute lung injury. Methods We co-cultured mouse lung microvascular endothelial cells (MLMVECs) with M2 macrophages following LPS challenge. M2 macrophages were intratracheally transplanted into mice subjected to cecal ligation and puncture (CLP). We further performed cytokine array for the supernatant from M2 macrophages and serum from mice subjected with CLP. Results We found both co-culture with M2 macrophages and treating with supernatant from M2 macrophages increased ECs viability following LPS challenge. Intratracheal transplantation of M2 macrophages markedly promoted pulmonary ECs proliferation, manifesting as attenuation of lung microvascular permeability and lung tissue edema, as well as improvement of survival rate. We further found that CXCL12, IL-1ra, TIMP-1, IL-4, and CXCL1 were increased in the supernatant of M2 macrophages in vitro. G-CSF and Complement Component 5a (C5/C5a) were increased in the serum of the M2-transplanted mice. Conclusions The present study suggested M2 macrophages could promote ECs proliferation in sepsis-induced ALI through secretion of anti-inflammatory cytokines and growth factors.
Collapse
Affiliation(s)
- Yaxing Shen
- Department of Thoracic Surgery, Zhongshan Hospital Fudan University, Shanghai 200032, China.,Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jieqiong Song
- Department of Critical Care Medicine, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Yingqin Wang
- Department of Critical Care Medicine, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Zhenglong Chen
- School of Medical Instrumentation, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
| | - Lin Zhang
- Center of Emergency and Intensive Care Unit, Jinshan Hospital Fudan University, Shanghai 201508, China
| | - Jie Yu
- Department of Critical Care Medicine, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Duming Zhu
- Department of Critical Care Medicine, Zhongshan Hospital Fudan University, Shanghai 200032, China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital Fudan University, Shanghai 200032, China
| |
Collapse
|
405
|
Zhang S, Ren Q, Qi H, Liu S, Liu Y. Adverse Effects of Fine-Particle Exposure on Joints and Their Surrounding Cells and Microenvironment. ACS NANO 2019; 13:2729-2748. [PMID: 30773006 DOI: 10.1021/acsnano.8b08517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Current understanding of the health risks and adverse effects upon exposure to fine particles is premised on the direct association of particles with target organs, particularly the lung; however, fine-particle exposure has also been found to have detrimental effects on sealed cavities distant to the portal-of-entry, such as joints. Moreover, the fundamental toxicological issues have been ascribed to the direct toxic mechanisms, in particular, oxidative stress and proinflammatory responses, without exploring the indirect mechanisms, such as compensated, adaptive, and secondary effects. In this Review, we recapitulate the current findings regarding the detrimental effects of fine-particle exposure on joints, the surrounding cells, and microenvironment, as well as their deteriorating impact on the progression of arthritis. We also elaborate the likely molecular mechanisms underlying the particle-induced detrimental influence on joints, not limited to direct toxicity, but also considering the other indirect mechanisms. Because of the similarities between fine air particles and engineered nanomaterials, we compare the toxicities of engineered nanomaterials to those of fine air particles. Arthritis and joint injuries are prevalent, particularly in the elderly population. Considering the severity of global exposure to fine particles and limited studies assessing the detrimental effects of fine-particle exposure on joints and arthritis, this Review aims to appeal to a broad interest and to promote more research efforts in this field.
Collapse
Affiliation(s)
- Shuping Zhang
- Institute for Medical Engineering and Science , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Quanzhong Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology , Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences , Beijing 100085 , P. R. China
| | - Hui Qi
- Beijing Jishuitan Hospital , Peking University Health Science Center , Beijing 100035 , P. R. China
- Beijing Research Institute of Traumatology and Orthopaedics , Beijing 100035 , P. R. China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology , Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences , Beijing 100085 , P. R. China
| | - Yajun Liu
- Beijing Jishuitan Hospital , Peking University Health Science Center , Beijing 100035 , P. R. China
| |
Collapse
|
406
|
Siouti E, Andreakos E. The many facets of macrophages in rheumatoid arthritis. Biochem Pharmacol 2019; 165:152-169. [PMID: 30910693 DOI: 10.1016/j.bcp.2019.03.029] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/19/2019] [Indexed: 12/24/2022]
Abstract
Macrophages are central to the pathophysiology of rheumatoid arthritis (RA). They constitute the main source of pro-inflammatory cytokines and chemokines such as TNF and IL-1β, they activate a wide range of immune and non-immune cells, and they secrete diverse tissue degrading enzymes driving chronic pro-inflammatory, tissue destructive and pain responses in RA. However, they can also produce anti-inflammatory cytokines such as IL-10, secrete inhibitors of tissue degrading enzymes and promote immunoregulatory and protective responses, suggesting the existence of macrophages with distinct and diverse functional activities. Although the underlying basis of this phenomenon has remained obscure for years, emerging evidence has now provided insight into the mechanisms and molecular processes involved. Here, we review current knowledge on the biology of macrophages in RA, and highlight recent literature on the heterogeneity, origins and ontogeny of macrophages as part of the mononuclear phagocyte system. We also discuss their plasticity in the context of the M1/M2 paradigm, and the emerging theme of metabolic rewiring as a major mechanism for programming macrophage functions and pro-inflammatory activities. This sheds light into the many facets of macrophages in RA, their molecular regulation and their translational potential for developing novel protective and therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Eleni Siouti
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Evangelos Andreakos
- Laboratory of Immunobiology, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; Airway Disease Infection Section, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London W2 1NY, United Kingdom.
| |
Collapse
|
407
|
Chan MWY, Viswanathan S. Recent progress on developing exogenous monocyte/macrophage-based therapies for inflammatory and degenerative diseases. Cytotherapy 2019; 21:393-415. [PMID: 30871899 DOI: 10.1016/j.jcyt.2019.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 02/04/2019] [Accepted: 02/13/2019] [Indexed: 12/14/2022]
Abstract
Cell-based therapies are a rapidly developing area of regenerative medicine as dynamic treatments that execute therapeutic functions multimodally. Monocytes and macrophages, as innate immune cells that control inflammation and tissue repair, are increasing popular clinical candidates due to their spectrum of functionality. In this article, we review the role of monocytes and macrophages specifically in inflammatory and degenerative disease pathology and the evidence supporting the use of these cells as an effective therapeutic strategy. We compare current strategies of exogenously polarized monocyte/macrophage therapies regarding dosage, delivery and processing to identify outcomes, advances and challenges to their clinical use. Monocytes/macrophages hold the potential to be a promising therapeutic avenue but understanding and optimization of disease-specific efficacy is needed to accelerate their clinical use.
Collapse
Affiliation(s)
- Mable Wing Yan Chan
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Sowmya Viswanathan
- Arthritis Program, Krembil Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Cell Therapy Program, University Health Network, Toronto, Ontario, Canada; Division of Hematology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
408
|
Das M, Laha D, Kanji S, Joseph M, Aggarwal R, Iwenofu OH, Pompili VJ, Jain MK, Das H. Induction of Krüppel-like factor 2 reduces K/BxN serum-induced arthritis. J Cell Mol Med 2019; 23:1386-1395. [PMID: 30506878 PMCID: PMC6349180 DOI: 10.1111/jcmm.14041] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 09/18/2018] [Accepted: 10/29/2018] [Indexed: 12/27/2022] Open
Abstract
Krüppel-like factor 2 (KLF2) critically regulates activation and function of monocyte, which plays important pathogenic role in progressive joint destruction in rheumatoid arthritis (RA). It is yet to be established the molecular basis of KLF2-mediated regulation of monocytes in RA pathogenesis. Herein, we show that a class of compound, HDAC inhibitors (HDACi) induced KLF2 expression in monocytes both in vitro and in vivo. KLF2 level was also elevated in tissues, such as bone marrow, spleen and thymus in mice after infusion of HDACi. Importantly, HDACi significantly reduced osteoclastic differentiation of monocytes with the up-regulation of KLF2 and concomitant down-regulation of matrixmetalloproteinases both in the expression level as well as in the protein level. In addition, HDACi reduced K/BxN serum-induced arthritic inflammation and joint destruction in mice in a dose-dependent manner. Finally, co-immunoprecipitation and overexpression studies confirmed that KLF2 directly interacts with HDAC4 molecule in cells. These findings provide mechanistic evidence of KLF2-mediated regulation of K/BxN serum-induced arthritic inflammation.
Collapse
Affiliation(s)
- Manjusri Das
- Department of Internal MedicineThe Ohio State University Medical CenterColumbusOhio
| | - Dipranjan Laha
- Department of Pharmaceutical SciencesSchool of PharmacyTexas Tech University Health Sciences CenterAmarilloTexas
| | - Suman Kanji
- Department of Pharmaceutical SciencesSchool of PharmacyTexas Tech University Health Sciences CenterAmarilloTexas
| | - Matthew Joseph
- Department of Internal MedicineThe Ohio State University Medical CenterColumbusOhio
| | - Reeva Aggarwal
- Department of Internal MedicineThe Ohio State University Medical CenterColumbusOhio
| | - Obiajulu H. Iwenofu
- Department of PathologyCollege of MedicineThe Ohio State UniversityColumbusOhio
| | - Vincent J. Pompili
- Department of Internal MedicineThe Ohio State University Medical CenterColumbusOhio
| | - Mukesh K. Jain
- Department of Internal MedicineCase Western Reserve UniversityClevelandOhio
| | - Hiranmoy Das
- Department of Pharmaceutical SciencesSchool of PharmacyTexas Tech University Health Sciences CenterAmarilloTexas
| |
Collapse
|
409
|
Chandrupatla DMSH, Molthoff CFM, Lammertsma AA, van der Laken CJ, Jansen G. The folate receptor β as a macrophage-mediated imaging and therapeutic target in rheumatoid arthritis. Drug Deliv Transl Res 2019; 9:366-378. [PMID: 30280318 PMCID: PMC6328514 DOI: 10.1007/s13346-018-0589-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Macrophages play a key role in the pathophysiology of rheumatoid arthritis (RA). Notably, positive correlations have been reported between synovial macrophage infiltration and disease activity as well as therapy outcome in RA patients. Hence, macrophages can serve as an important target for both imaging disease activity and drug delivery in RA. Folate receptor β (FRβ) is a glycosylphosphatidyl (GPI)-anchored plasma membrane protein being expressed on myeloid cells and activated macrophages. FRβ harbors a nanomolar binding affinity for folic acid allowing this receptor to be exploited for RA disease imaging (e.g., folate-conjugated PET tracers) and therapeutic targeting (e.g., folate antagonists and folate-conjugated drugs). This review provides an overview of these emerging applications in RA by summarizing and discussing properties of FRβ, expression of FRβ in relation to macrophage polarization, FRβ-targeted in vivo imaging modalities, and FRβ-directed drug targeting.
Collapse
Affiliation(s)
- Durga M S H Chandrupatla
- Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Carla F M Molthoff
- Department of Radiology and Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Conny J van der Laken
- Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Gerrit Jansen
- Amsterdam Rheumatology and Immunology Center, VU University Medical Center, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
410
|
Guan L, Yu D, Wu GH, Ning HJ, He SD, Li SS, Hu TY, Yang G, Liu ZQ, Yu HQ, Sun XZ, Liu ZG, Yang PC. Vasoactive intestinal peptide is required in the maintenance of immune regulatory competency of immune regulatory monocytes. Clin Exp Immunol 2019; 196:276-286. [PMID: 30636174 DOI: 10.1111/cei.13259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2019] [Indexed: 11/27/2022] Open
Abstract
Dysfunction of the immune regulatory system plays an important role in the pathogenesis of rheumatoid arthritis (RA). Vasoactive intestinal peptide (VIP) has multiple bioactivities. This study aims to investigate the role of VIP in the maintenance of the immune regulatory capacity of monocytes (Mos). Human peripheral blood samples were collected from RA patients and healthy control (HC) subjects. Mos and CD14+ CD71- CD73+ CD25+ regulatory Mos (RegMos) were isolated from the blood samples and characterized by flow cytometry. A rat RA model was developed to test the role of VIP in the maintenance of the immune regulatory function of Mos. The results showed that RegMos of HC subjects had immune suppressive functions. RegMos of RA patients expressed less interleukin (IL)-10 and showed an incompetent immune regulatory capacity. Serum levels of VIP were lower in RA patients, which were positively correlated with the expression of IL-10 in RegMos. In-vitro experiments showed that the IL-10 mRNA decayed spontaneously in RegMos, which could be prevented by the presence of VIP in the culture. VIP suppressed the effects of tristetraprolin (TTP) on inducing IL-10 mRNA decay in RegMos. Administration of VIP inhibited experimental RA in rats through restoring the IL-10 expression in RegMos. RegMos have immune suppressive functions. VIP is required in maintaining IL-10 expression in RegMos. The data suggest that VIP has translational potential in the treatment of immune disorders such as RA.
Collapse
Affiliation(s)
- L Guan
- Department of Respirology, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - D Yu
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - G-H Wu
- Department of Respirology, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - H-J Ning
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - S-D He
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - S-S Li
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - T-Y Hu
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Longgang ENT Hospital and Shenzhen ENT Institute, Shenzhen, China
| | - G Yang
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Longgang ENT Hospital and Shenzhen ENT Institute, Shenzhen, China
| | - Z-Q Liu
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Longgang ENT Hospital and Shenzhen ENT Institute, Shenzhen, China
| | - H-Q Yu
- Department of Respirology, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - X-Z Sun
- Department of Respirology, Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Z-G Liu
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - P-C Yang
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
411
|
Koh JH, Park YJ, Lee S, Hong YS, Hong KS, Yoo SA, Cho CS, Kim WU. Distinct Urinary Metabolic Profile in Rheumatoid Arthritis Patients: A Possible Link between Diet and Arthritis Phenotype. JOURNAL OF RHEUMATIC DISEASES 2019. [DOI: 10.4078/jrd.2019.26.1.46] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Jung Hee Koh
- Division of Rheumatology, Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Yune-Jung Park
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Saseong Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Korea
| | - Young-Shick Hong
- Division of Food and Nutrition, Chonnam National University, Gwangju, Korea
| | - Kwan Soo Hong
- Bioimaging Research Team, Korea Basic Science Institute, Cheongju, Korea
| | - Seung-Ah Yoo
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Korea
| | - Chul-Soo Cho
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Korea
| | - Wan-Uk Kim
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
412
|
Moise N, Friedman A. Rheumatoid arthritis - a mathematical model. J Theor Biol 2019; 461:17-33. [DOI: 10.1016/j.jtbi.2018.10.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/17/2018] [Accepted: 10/18/2018] [Indexed: 12/18/2022]
|
413
|
Xu X, Xu J, Wu J, Hu Y, Han Y, Gu Y, Zhao K, Zhang Q, Liu X, Liu J, Liu B, Cao X. Phosphorylation-Mediated IFN-γR2 Membrane Translocation Is Required to Activate Macrophage Innate Response. Cell 2018; 175:1336-1351.e17. [PMID: 30318148 DOI: 10.1016/j.cell.2018.09.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 08/04/2018] [Accepted: 09/07/2018] [Indexed: 01/20/2023]
Abstract
As a critical step during innate response, the cytoplasmic β subunit (IFN-γR2) of interferon-γ receptor (IFN-γR) is induced and translocates to plasma membrane to join α subunit to form functional IFN-γR to mediate IFN-γ signaling. However, the mechanism driving membrane translocation and its significance remain largely unknown. We found, unexpectedly, that mice deficient in E-selectin, an endothelial cell-specific adhesion molecule, displayed impaired innate activation of macrophages upon Listeria monocytogenes infection yet had increased circulating IFN-γ. Inflammatory macrophages from E-selectin-deficient mice had less surface IFN-γR2 and impaired IFN-γ signaling. BTK elicited by extrinsic E-selectin engagement phosphorylates cytoplasmic IFN-γR2, facilitating EFhd2 binding and promoting IFN-γR2 trafficking from Golgi to cell membrane. Our findings demonstrate that membrane translocation of cytoplasmic IFN-γR2 is required to activate macrophage innate response against intracellular bacterial infection, identifying the assembly of functional cytokine receptors on cell membrane as an important layer in innate activation and cytokine signaling.
Collapse
Affiliation(s)
- Xiaoqing Xu
- Department of Immunology and Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, 100005 Beijing, China; National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, 200433 Shanghai, China
| | - Jia Xu
- Department of Immunology and Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Jiacheng Wu
- Department of Immunology and Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Ye Hu
- Department of Immunology and Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Yanmei Han
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, 200433 Shanghai, China
| | - Yan Gu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, 200433 Shanghai, China
| | - Kai Zhao
- Department of Immunology and Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, 100005 Beijing, China
| | - Qian Zhang
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, 200433 Shanghai, China
| | - Xingguang Liu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, 200433 Shanghai, China
| | - Juan Liu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, 200433 Shanghai, China
| | - Bing Liu
- Translational Medicine Center, Academy of Military Medical Sciences, 100024 Beijing, China
| | - Xuetao Cao
- Department of Immunology and Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, 100005 Beijing, China; National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, 200433 Shanghai, China; College of Life Science, Nankai University, 300071 Tianjin, China.
| |
Collapse
|
414
|
Woo SJ, Noh HS, Lee NY, Cheon YH, Yi SM, Jeon HM, Bae EJ, Lee SI, Park BH. Myeloid sirtuin 6 deficiency accelerates experimental rheumatoid arthritis by enhancing macrophage activation and infiltration into synovium. EBioMedicine 2018; 38:228-237. [PMID: 30429089 PMCID: PMC6306347 DOI: 10.1016/j.ebiom.2018.11.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/23/2018] [Accepted: 11/02/2018] [Indexed: 11/28/2022] Open
Abstract
Background We recently reported that myeloid sirtuin 6 (Sirt6) is a critical determinant of phenotypic switching and the migratory responses of macrophages. Given the prominent role of macrophages in the pathogenesis of rheumatoid arthritis (RA), we tested whether myeloid Sirt6 deficiency affects the development and exacerbation of RA. Methods Arthritis was induced in wild type and myeloid Sirt6 knockout (mS6KO) mice using collagen-induced and K/BxN serum transfer models. Sirt6 expression (or activity) and inflammatory activities were compared in peripheral blood mononuclear cells (PBMCs) and monocytes/macrophages obtained from patients with RA or osteoarthritis. Findings Based on clinical score, ankle thickness, pathology, and radiology, arthritis was more severe in mS6KO mice relative to wild type, with a greater accumulation of macrophages in the synovium. Consistent with these findings, myeloid Sirt6 deficiency increased the migration potential of macrophages toward synoviocyte-derived chemoattractants. Mechanistically, Sirt6 deficiency in macrophages caused an inflammation with increases in acetylation and protein stability of forkhead box protein O1. Conversely, ectopic overexpression of Sirt6 in knockout cells reduced the inflammatory responses. Lastly, PBMCs and monocytes/macrophages from RA patients exhibited lower expression of Sirt6 than those from patients with osteoarthritis, and their Sirt6 activity was inversely correlated with disease severity. Interpretation Our data identify a role of myeloid Sirt6 in clinical and experimental RA and suggest that myeloid Sirt6 may be an intriguing therapeutic target. Fund Medical Research Center Program and Basic Science Research Program through the National Research Foundation of Korea. Myeloid Sirt6 deficiency aggravates the joint destruction by increasing recruitment of macrophages into arthritic joints. Myeloid Sirt6 deacetylates FoxO1 to promote proteasomal degradation. Overexpression of Sirt6 greatly attenuates inflammatory activity of human macrophages. Sirt6 expression and activity decrease in blood monocytes and joint macrophages from RA patients.
Collapse
Affiliation(s)
- Seong Ji Woo
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Hae Sook Noh
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Gyeongnam 52828, Republic of Korea
| | - Na Young Lee
- Department of Internal Medicine, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea
| | - Yun-Hong Cheon
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Gyeongnam 52828, Republic of Korea
| | - Sang Mi Yi
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Gyeongnam 52828, Republic of Korea
| | - Hyun Min Jeon
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Gyeongnam 52828, Republic of Korea
| | - Eun Ju Bae
- College of Pharmacy, Woosuk University, Wanju, Jeonbuk 55338, Republic of Korea
| | - Sang-Il Lee
- Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Gyeongnam 52828, Republic of Korea.
| | - Byung-Hyun Park
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju, Jeonbuk 54896, Republic of Korea.
| |
Collapse
|
415
|
Lin R, Liu Y, Piao M, Song Y. Magnesium isoglycyrrhizinate positively affects concanavalin A-induced liver damage by regulating macrophage polarization. FOOD AGR IMMUNOL 2018. [DOI: 10.1080/09540105.2018.1508424] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Rui Lin
- Department of Gastroenterology and Hepatology, Tianjin Medical University, General Hospital, Tianjin, People’s Republic of China
| | - Yun Liu
- Tianjin Bonagene Bio-Technology Co. Ltd., Tianjin, People’s Republic of China
- Academician Workstation of Hunan Baodong Farming Co. Ltd., Hunan, People’s Republic of China
| | - Meiyu Piao
- Department of Gastroenterology and Hepatology, Tianjin Medical University, General Hospital, Tianjin, People’s Republic of China
| | - Yan Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University, General Hospital, Tianjin, People’s Republic of China
| |
Collapse
|
416
|
Lv L, Xie Y, Li K, Hu T, Lu X, Cao Y, Zheng X. Unveiling the Mechanism of Surface Hydrophilicity-Modulated Macrophage Polarization. Adv Healthc Mater 2018; 7:e1800675. [PMID: 30106513 DOI: 10.1002/adhm.201800675] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/24/2018] [Indexed: 12/22/2022]
Abstract
With inflammation increasingly recognized as a key factor that influences fracture healing, the immunologic response is considered to play a pivotal role in determining implant-mediated osteogenesis. Herein, this paper demonstrates that modification of the surface hydrophilicity of Ti surface oxides can be utilized to control immune response by steering the macrophage polarization toward pro- or anti-inflammation phenotype. Enhanced anti-inflammatory and prohealing performance of macrophages is observed on hydrophilic surfaces compared to hydrophobic ones. Further study on the detailed mechanism demonstrates that the surface hydrophilicity controls specific proteins (fibronectin and fibrinogen) adsorption and conformation, which activate different signaling pathways (PI3K and NF-κB) through selective expression of integrin β1 or β2 to influence the behaviors of macrophages. Thus, this study presents a mechanism of macrophage polarization modulated by surface hydrophilicity for the surface design of advanced implant materials with satisfactory anti-inflammatory and osteogenesis-promoting properties.
Collapse
Affiliation(s)
- Lin Lv
- Key Laboratory of Inorganic Coating Materials CASShanghai Institute of CeramicsChinese Academy of Sciences 1295 Dingxi Road Shanghai 200050 P. R. China
- University of Chinese Academy of Sciences 19 Yuquan Road Beijing 100049 P. R. China
| | - Youtao Xie
- Key Laboratory of Inorganic Coating Materials CASShanghai Institute of CeramicsChinese Academy of Sciences 1295 Dingxi Road Shanghai 200050 P. R. China
| | - Kai Li
- Key Laboratory of Inorganic Coating Materials CASShanghai Institute of CeramicsChinese Academy of Sciences 1295 Dingxi Road Shanghai 200050 P. R. China
| | - Tao Hu
- Department of Spine SurgeryShanghai East HospitalTongji University School of Medicine Shanghai 200050 P. R. China
| | - Xiang Lu
- Key Laboratory of Inorganic Coating Materials CASShanghai Institute of CeramicsChinese Academy of Sciences 1295 Dingxi Road Shanghai 200050 P. R. China
- University of Chinese Academy of Sciences 19 Yuquan Road Beijing 100049 P. R. China
| | - Yunzhen Cao
- Key Laboratory of Inorganic Coating Materials CASShanghai Institute of CeramicsChinese Academy of Sciences 1295 Dingxi Road Shanghai 200050 P. R. China
| | - Xuebin Zheng
- Key Laboratory of Inorganic Coating Materials CASShanghai Institute of CeramicsChinese Academy of Sciences 1295 Dingxi Road Shanghai 200050 P. R. China
| |
Collapse
|
417
|
Wang Q, Zhou X, Zhao Y, Xiao J, Lu Y, Shi Q, Wang Y, Wang H, Liang Q. Polyphyllin I Ameliorates Collagen-Induced Arthritis by Suppressing the Inflammation Response in Macrophages Through the NF-κB Pathway. Front Immunol 2018; 9:2091. [PMID: 30319603 PMCID: PMC6170622 DOI: 10.3389/fimmu.2018.02091] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 08/24/2018] [Indexed: 12/27/2022] Open
Abstract
Background: Rheumatoid arthritis (RA) is a chronic autoimmune disorder, characterized by an increased number of M1-like macrophages in the joints. Polyphyllin I (PPI), one of the main components in the Rhizoma of Paris polyphyllin, displays a selective inhibitory effect on various tumor cells. Here we sought to investigate the anti-rheumatoid arthritis effects and mechanisms of PPI on macrophages in vivo and in vitro. Materials and Methods:In vitro, primary bone marrow-derived macrophages (BMMs) and peritoneal elucidated macrophages (PEMs) were stimulated by lipopolysaccharide (LPS) and Interferon (IFN)-γ and then treated with PPI. We determined the degree of activation of IKKα/β and p65, two key mediators of the NF-κB-mediated inflammatory pathway, by measuring their phosphorylated forms by Western blot. The p65 nuclear localization was detected by immunofluorescent staining. Further, a NF-κB-linked luciferase reporter plasmid, as well as those expressing key mediators of the Toll-like receptor 4 pathway, such as myeloid differentiation primary response 88 (MYD88), interleukin-1 receptor (IL-1R) associated kinase (IRAK)-1, TNF receptor associated factors (TRAF)-6, Transforming growth factor-b–activated kinase 1 (TAK1) and p65, were used to identify the mechanism by which PPI achieves its inhibitory effects on macrophage-mediated inflammation. Moreover, a NF-κB inhibitor, p65-targeted siRNAs, and a p65 plasmid were further used to validate the anti-inflammatory mechanism of PPI. In vivo, PPI (1 mg/kg) was administered intragastrically one time a day for 7 weeks starting on the 42nd day after the first immunization with collagen in a collagen-induced arthritis (CIA) mouse model. Micro-computed Tomography scanning, histological examination, F4/80 and iNOS double immunofluorescent staining and CD4 immunohistochemical staining were performed to determine the effect of PPI treatment on joint structure and inflammation in this model. Results: PPI reduced the inflammatory cytokines production of PEMs stimulated by LPS/IFN-γ, inhibited the phosphorylation of IKKα/β and p65, and prevented p65 nuclear localization. The NF-κB luciferase assay showed that the target of PPI was closely related to the NF-κB pathway. Moreover, NF-κB inhibition, siRNA-mediated knockdown of p65, and p65 overexpression eliminated PPI's inhibitory effect. In addition, PPI attenuated the bone erosion and synovitis, as well as M1-like macrophage and T cell infiltration, in the ankle joint of the CIA model. Conclusion: PPI demonstrated effective amelioration of synovial inflammation in the ankle joint of CIA mice while suppressing NF-κB-mediated production of pro-inflammatory effectors in activated macrophages.
Collapse
Affiliation(s)
- Qiong Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,State Key Laboratory of Cell Biology, Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Innovation Center for Cell Signaling Network, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin Zhou
- State Key Laboratory of Cell Biology, Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Innovation Center for Cell Signaling Network, Shanghai, China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Jun Xiao
- State Key Laboratory of Cell Biology, Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Innovation Center for Cell Signaling Network, Shanghai, China
| | - Yao Lu
- State Key Laboratory of Cell Biology, Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Innovation Center for Cell Signaling Network, Shanghai, China
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China.,School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongyan Wang
- State Key Laboratory of Cell Biology, Key Laboratory of Systems Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Innovation Center for Cell Signaling Network, Shanghai, China
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Spine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education (Shanghai University of Traditional Chinese Medicine), Shanghai, China
| |
Collapse
|
418
|
Liu W, Zhang Y, Zhu W, Ma C, Ruan J, Long H, Wang Y. Sinomenine Inhibits the Progression of Rheumatoid Arthritis by Regulating the Secretion of Inflammatory Cytokines and Monocyte/Macrophage Subsets. Front Immunol 2018; 9:2228. [PMID: 30319663 PMCID: PMC6168735 DOI: 10.3389/fimmu.2018.02228] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/07/2018] [Indexed: 12/26/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory arthropathy associated with articular damage and attendant comorbidities. Even although RA treatment has advanced remarkably over the last decade, a significant proportion of patients still do not achieve sustained remission. The cause of RA is not yet known despite the many potential mechanisms proposed. It has been confirmed that RA is associated with dysregulated immune system and persistent inflammation. Therefore, management of inflammation is always the target of therapy. Sinomenine (SIN) is the prescription drug approved by the Chinese government for RA treatment. A previous study found that SIN was a robust anti-inflammation drug. In this study, we screened the different secretory cytokines using inflammation antibody arrays and qRT-PCR in both LPS-induced and SIN-treated RAW264.7 cells followed by evaluation of the ability of SIN to modulate cytokine secretion in a cell model, collagen-induced arthritis (CIA) mouse model, and RA patients. Several clinical indexes affecting the 28-joint disease activity score (DAS28) were determined before and after SIN treatment. Clinical indexes, inflammatory cytokine secretion, and DAS28 were compared among RA patients treated with either SIN or methotrexate (MTX). To explore the mechanism of SIN anti-inflammatory function, RA-associated monocyte/macrophage subsets were determined using flow cytometry in CIA mouse model and RA patients, both treated with SIN. The results demonstrated that SIN regulated IL-6, GM-CSF, IL-12 p40, IL-1α, TNF-α, IL-1β, KC (CXCL1), Eotaxin-2, IL-10, M-CSF, RANTES, and MCP-1 secretion in vivo and in vitro and reduced RA activity and DAS28 in a clinical setting. Furthermore, SIN attenuated CD11b+F4/80+CD64+ resident macrophages in the synovial tissue, CD11b+Ly6C+CD43+ macrophages in the spleen and draining lymph nodes of CIA mice. The percentage of CD14+CD16+ peripheral blood mononuclear cells was reduced by SIN in RA patients. These data indicated that SIN regulates the secretion of multiple inflammatory cytokines and monocyte/macrophage subsets, thereby suppressing RA progression. Therefore, along with MTX, SIN could be an alternative cost-effective anti-inflammatory agent for treating RA.
Collapse
Affiliation(s)
- Weiwei Liu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Weina Zhu
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunhua Ma
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Ruan
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongyan Long
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pediatrics, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Wang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China.,The First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
419
|
Sendo S, Saegusa J, Morinobu A. Myeloid-derived suppressor cells in non-neoplastic inflamed organs. Inflamm Regen 2018; 38:19. [PMID: 30237829 PMCID: PMC6139938 DOI: 10.1186/s41232-018-0076-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Myeloid-derived suppressor cells (MDSCs) are a highly heterogeneous population of immature myeloid cells with immunosuppressive function. Although their function in tumor-bearing conditions is well studied, less is known about the role of MDSCs in various organs under non-neoplastic inflammatory conditions. MAIN BODY MDSCs are divided into two subpopulations, G-MDSCs and M-MDSCs, and their distribution varies between organs. MDSCs negatively control inflammation in inflamed organs such as the lungs, joints, liver, kidneys, intestines, central nervous system (CNS), and eyes by suppressing T cells and myeloid cells. MDSCs also regulate fibrosis in the lungs, liver, and kidneys and help repair CNS injuries. MDSCs in organs are plastic and can differentiate into osteoclasts and tolerogenic dendritic cells according to the microenvironment under non-neoplastic inflammatory conditions. CONCLUSION This article summarizes recent findings about MDSCs under inflammatory conditions, especially with respect to their function and differentiation in specific organs.
Collapse
Affiliation(s)
- Sho Sendo
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| | - Jun Saegusa
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
- Division of Laboratory Medicine, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| | - Akio Morinobu
- Division of Rheumatology and Clinical Immunology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017 Japan
| |
Collapse
|
420
|
Swart M, Troeberg L. Effect of Polarization and Chronic Inflammation on Macrophage Expression of Heparan Sulfate Proteoglycans and Biosynthesis Enzymes. J Histochem Cytochem 2018; 67:9-27. [PMID: 30205019 DOI: 10.1369/0022155418798770] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Heparan sulfate (HS) proteoglycans on immune cells have the ability to bind to and regulate the bioactivity more than 400 bioactive protein ligands, including many chemokines, cytokines, and growth factors. This makes them important regulators of the phenotype and behavior of immune cells. Here we review how HS biosynthesis in macrophages is regulated during polarization and in chronic inflammatory diseases such as rheumatoid arthritis, atherosclerosis, asthma, chronic obstructive pulmonary disease and obesity, by analyzing published micro-array data and mechanistic studies in this area. We describe that macrophage expression of many HS biosynthesis and core proteins is strongly regulated by macrophage polarization, and that these expression patterns are recapitulated in chronic inflammation. Such changes in HS biosynthetic enzyme expression are likely to have a significant impact on the phenotype of macrophages in chronic inflammatory diseases by altering their interactions with chemokines, cytokines, and growth factors.
Collapse
Affiliation(s)
- Maarten Swart
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Linda Troeberg
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
421
|
Chandrupatla DMSH, Molthoff CFM, Ritsema WIGR, Vos R, Elshof E, Matsuyama T, Low PS, Musters RJP, Hammond A, Windhorst AD, Lammertsma AA, van der Laken CJ, Brands R, Jansen G. Prophylactic and therapeutic activity of alkaline phosphatase in arthritic rats: single-agent effects of alkaline phosphatase and synergistic effects in combination with methotrexate. Transl Res 2018; 199:24-38. [PMID: 29802817 DOI: 10.1016/j.trsl.2018.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/09/2018] [Accepted: 04/03/2018] [Indexed: 12/16/2022]
Abstract
Alkaline phosphatase (AP) is a gate-keeper of innate immune system responses by detoxifying inflammation triggering moieties released from endogenous and external sources. We examined whether AP's broad mechanism of action constitutes a safe therapeutic, either as single agent or combined with methotrexate (MTX), for chronic inflammatory disorders, for example, rheumatoid arthritis (RA). A rat model for RA was used with repeated intra-articular methylated bovine serum albumin (mBSA) injections in 1 knee ("arthritic" knee), with the contralateral knee serving as internal control. AP (200 µg, subcut) was administered before mBSA injections (prophylactic setting) or after arthritis induction (therapeutic setting) or combined with MTX (0.3 mg/kg or 1 mg/kg; intraperitoneally). As end point of treatment outcome, macrophage infiltration in knees, liver, and spleen was assessed by immunohistochemistry (ED1 and ED2 expression), immunofluoresence (macrophage marker folate receptor-β [FRβ]), and [18F]fluoro-polyethylene glycol-folate positron emission tomography (PET) (macrophage imaging) and ex vivo tissue distribution. Single-agent AP treatment and combinations with MTX were well tolerated. Both prophylactic and therapeutic AP markedly reduced synovial macrophage infiltration in arthritic knees (ED1: 3.5- to 4-fold; ED2: 3.5- to 6-fold), comparable with MTX treatment. AP-MTX combinations slightly improved on single agent effects. PET monitoring and ex vivo tissue distribution studies corroborated the impact of AP, MTX, and AP-MTX on reducing synovial macrophage infiltration. Beyond localized articular effects, AP also revealed systemic anti-inflammatory effects by a 2-fold reduction of ED1, ED2, and FRβ+ macrophages in liver and spleen of arthritic rats. Collectively, single-agent AP and AP combined with MTX elicited local and systemic anti-arthritic activity in arthritic rats.
Collapse
Affiliation(s)
- Durga M S H Chandrupatla
- Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | - Carla F M Molthoff
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Wayne I G R Ritsema
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Ricardo Vos
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Eline Elshof
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Takami Matsuyama
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - René J P Musters
- Department of Physiology, VU University Medical Center, Amsterdam, The Netherlands
| | - Anthony Hammond
- Department of Rheumatology, KIMS Hospital, Kent, United Kingdom
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Conny J van der Laken
- Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, The Netherlands
| | | | - Gerrit Jansen
- Amsterdam Rheumatology and immunology Center, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
422
|
Luteolin Induces Apoptosis and Autophagy in Mouse Macrophage ANA-1 Cells via the Bcl-2 Pathway. J Immunol Res 2018; 2018:4623919. [PMID: 30246034 PMCID: PMC6136535 DOI: 10.1155/2018/4623919] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/05/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Abstract
Plants rich in luteolin have been used as Chinese traditional medicines for inflammatory diseases, hypertension, and cancer. However, little is known about the effect of luteolin on the apoptosis or autophagy of the macrophages. In this study, mouse macrophage ANA-1 cells were incubated with different concentrations of luteolin. The viability of the cells was determined by an MTT assay, apoptosis was determined by flow cytometric analysis, the level of cell autophagy was observed by confocal microscopy, and the expression levels of apoptotic or autophagic and antiapoptotic or antiautophagic proteins were detected by Western blot analysis. The results showed that luteolin decreased the viability of ANA-1 cells and induced apoptosis and autophagy. Luteolin induced apoptosis accompanied by downregulation of the expression of Bcl-2 and upregulation of the expression of caspase 3 and caspase 8. And luteolin increased FITC-LC3 punctate fluorescence accompanied by the increased expression levels of LC3-I, ATG7, and ATG12, while it suppressed the expression level of Beclin-1. Luteolin treatment resulted in obvious activation of the p38, JNK, and Akt signaling pathways, which is important in modulating apoptosis and autophagy. Thus, we concluded that luteolin induced the apoptosis and autophagy of ANA-1 cells most likely by regulating the p38, JNK, and Akt pathways, inhibiting the activity of Bcl-2 and Beclin-1 and upregulating caspase 3 and caspase 8 expression. These results provide novel insights into a therapeutic strategy to prevent and possibly treat macrophage-related diseases through luteolin-induced apoptosis and autophagy.
Collapse
|
423
|
Jia X, Hu X, Han S, Miao X, Liu H, Li X, Lin Z, Wang Z, Gong W. Increased M1 macrophages in young miR-15a/16−/−
mice with tumour grafts or dextran sulphate sodium-induced colitis. Scand J Immunol 2018; 88:e12703. [DOI: 10.1111/sji.12703] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/12/2018] [Indexed: 02/07/2023]
Affiliation(s)
- Xiaoqin Jia
- Department of Pathology; Institute of Translational Medicine; Medical College; Yangzhou University; Yangzhou China
| | - Xiangyu Hu
- Department of Pathology; Institute of Translational Medicine; Medical College; Yangzhou University; Yangzhou China
| | - Sen Han
- Department of Pathology; Institute of Translational Medicine; Medical College; Yangzhou University; Yangzhou China
| | - Xin Miao
- Department of Pathology; Institute of Translational Medicine; Medical College; Yangzhou University; Yangzhou China
| | - Hao Liu
- Department of General Surgery; Subei People's Hospital of Jiangsu Province; Yangzhou University; Yangzhou China
| | - Xiaomin Li
- Department of Pathology; Institute of Translational Medicine; Medical College; Yangzhou University; Yangzhou China
| | - Zhijie Lin
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research; Yangzhou University; Yangzhou China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases; Yangzhou University; Yangzhou China
- Jiangsu Key Laboratory of Zoonosis; Yangzhou University; Yangzhou China
| | - Zhengbing Wang
- Department of General Surgery; Subei People's Hospital of Jiangsu Province; Yangzhou University; Yangzhou China
| | - Weijuan Gong
- Department of Pathology; Institute of Translational Medicine; Medical College; Yangzhou University; Yangzhou China
- Department of General Surgery; Subei People's Hospital of Jiangsu Province; Yangzhou University; Yangzhou China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research; Yangzhou University; Yangzhou China
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases; Yangzhou University; Yangzhou China
- Jiangsu Key Laboratory of Zoonosis; Yangzhou University; Yangzhou China
| |
Collapse
|
424
|
Reduction of Articular and Systemic Inflammation by Kava-241 in a Porphyromonas gingivalis-Induced Arthritis Murine Model. Infect Immun 2018; 86:IAI.00356-18. [PMID: 29914930 DOI: 10.1128/iai.00356-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory disease that has been linked to several risk factors, including periodontitis. Identification of new anti-inflammatory compounds to treat arthritis is needed. We had previously demonstrated the beneficial effect of Kava-241, a kavain-derived compound, in the management of Porphyromonas gingivalis-induced periodontitis. The present study evaluated systemic and articular effects of Kava-241 in an infective arthritis murine model triggered by P. gingivalis bacterial inoculation and primed with a collagen antibody cocktail (CIA) to induce joint inflammation and tissular destruction. Clinical inflammation score and radiological analyses of the paws were performed continuously, while histological assessment was obtained at sacrifice. Mice exposed to P. gingivalis and a CIA cocktail and treated concomitantly with Kava-241 exhibited a reduced clinical inflammatory score and a decreased number of inflammatory cells and osteoclasts within joint. Kava-241 treatment also decreased significantly tumor necrosis factor alpha (TNF-α) in serum from mice injected with a Toll-like receptor 2 or 4 (TLR-2/4) ligand, P. gingivalis-lipopolysaccharide (LPS). Finally, bone marrow-derived macrophages infected with P. gingivalis and exposed to Kava-241 displayed reduced TLR-2/4, reduced mitogen-activated protein kinase (MAPK)-related signal elements, and reduced LPS-induced TNF-α factor (LITAF), all explaining the observed reduction of TNF-α secretion. Taken together, these results emphasized the novel properties of Kava-241 in the management of inflammatory conditions, especially TNF-α-related diseases such as infective RA.
Collapse
|
425
|
Travelli C, Colombo G, Mola S, Genazzani AA, Porta C. NAMPT: A pleiotropic modulator of monocytes and macrophages. Pharmacol Res 2018; 135:25-36. [PMID: 30031171 DOI: 10.1016/j.phrs.2018.06.022] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 06/20/2018] [Indexed: 12/11/2022]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is the bottleneck enzyme of the NAD salvage pathway and thereby is a controller of intracellular NAD concentrations. It has been long known that the same enzyme can be secreted by a number of cell types and acts as a cytokine, although its receptor is at present unknown. Investigational compounds have been developed that target the enzymatic activity as well as the extracellular action (i.e. neutralizing antibodies). The present contribution reviews the evidence that links intracellular and extracellular NAMPT to myeloid biology, for example governing monocyte/macrophage differentiation, polarization and migration. Furthermore, it reviews the evidence that links this protein to some disorders in which myeloid cells have a prominent role (acute infarct, inflammatory bowel disease, acute lung injury and rheumatoid arthritis) and the data showing that inhibition of the enzymatic activity or the neutralization of the cytokine is beneficial in preclinical animal models.
Collapse
Affiliation(s)
- Cristina Travelli
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Giorgia Colombo
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Silvia Mola
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy.
| | - Chiara Porta
- Department of Pharmacological Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
426
|
Ma J, Guo A, Wang S, Man S, Zhang Y, Liu S, Liu Y. From the lung to the knee joint: Toxicity evaluation of carbon black nanoparticles on macrophages and chondrocytes. JOURNAL OF HAZARDOUS MATERIALS 2018; 353:329-339. [PMID: 29680691 DOI: 10.1016/j.jhazmat.2018.04.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/11/2018] [Accepted: 04/12/2018] [Indexed: 06/08/2023]
Abstract
Carbon black (CB), a core elemental carbon component of airborne particles, has been used as a model material to study environmental safety and health impacts of airborne particles. Although potential adverse effects of CB have been reported, limited knowledge is available regarding CB-induced metabolic disorders and secondary effects distant from primary target organs, such as the effects on joints. The knee cavity is a relatively closed space along the peripheral circulation route with a slow rate of interchange of nutrition with blood. While epidemiologic studies have indicated that airborne particle exposure may affect the occurrence and severity of inflammatory knee diseases, no research has been performed to understand the potential hazardous direct/indirect interactions between particles and knee cells. Herein, we have scrutinized the toxicity of four commercial nano-sized CB samples in the lung and a distant site: knee joint. Our results indicated that CB triggered pulmonary and systemic inflammation upon inhalation exposure, and, more strikingly, CB also elicited injuries of the knee joint, as demonstrated by thickened synovial membrane, suggesting disordered cellular metabolism within the knee joint. Our data recognized the CB toxicity profiles to macrophages as characterized by pro-inflammatory reactions, and also defined an activated metabolic state of chondrocytes, as evidenced by metalloproteinase (MMP) induction. Of note, remarkable variations were also found for these changes induced by these four CB samples, due to their distinct physicochemical properties. Collectively, our results uncovered a significant toxicity of CB inhalation exposure to the knee joint, as reflected by metabolic activation of chondrocytes, and, more importantly, these findings unearthed CB-induced metabolic disorders and secondary effects owing to systemic pro-inflammatory conditions upon CB exposure, in addition to the likelihood of direct toxicity to knee cells.
Collapse
Affiliation(s)
- Juan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Anyi Guo
- Beijing Jishuitan Hospital, Peking University Health Science Center, Beijing, 100035, PR China
| | - Shunhao Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Siliang Man
- Beijing Jishuitan Hospital, Peking University Health Science Center, Beijing, 100035, PR China
| | - Yunjian Zhang
- Beijing Jishuitan Hospital, Peking University Health Science Center, Beijing, 100035, PR China
| | - Sijin Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, PR China; University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Yajun Liu
- Beijing Jishuitan Hospital, Peking University Health Science Center, Beijing, 100035, PR China.
| |
Collapse
|
427
|
Zhao HY, Lyu ZS, Duan CW, Song Y, Han TT, Mo XD, Wang Y, Xu LP, Zhang XH, Huang XJ, Kong Y. An unbalanced monocyte macrophage polarization in the bone marrow microenvironment of patients with poor graft function after allogeneic haematopoietic stem cell transplantation. Br J Haematol 2018; 182:679-692. [PMID: 29974948 DOI: 10.1111/bjh.15452] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/17/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Hong-Yan Zhao
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
| | - Zhong-Shi Lyu
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
- Peking-Tsinghua Center for Life Sciences; Academy for Advanced Interdisciplinary Studies; Peking University; Beijing China
| | - Cai-Wen Duan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health and Pediatric Translational Medicine Institute; Shanghai Children's Medical Center; Shanghai Collaborative Innovation Center for Translational Medicine and Department of Pharmacology and Chemical Biology; Shanghai Jiao Tong University School of medicine; Shanghai China
| | - Yang Song
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
- Peking-Tsinghua Center for Life Sciences; Academy for Advanced Interdisciplinary Studies; Peking University; Beijing China
| | - Ting-Ting Han
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
| | - Xiao-Dong Mo
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
| | - Yu Wang
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
| | - Lan-Ping Xu
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
| | - Xiao-Hui Zhang
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
| | - Xiao-Jun Huang
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
- Peking-Tsinghua Center for Life Sciences; Academy for Advanced Interdisciplinary Studies; Peking University; Beijing China
| | - Yuan Kong
- Peking University People's Hospital; Peking University Institute of Hematology; Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation; Collaborative Innovation Center of Hematology; Peking University; Beijing China
| |
Collapse
|
428
|
Targeting Macrophages as a Potential Therapeutic Intervention: Impact on Inflammatory Diseases and Cancer. Int J Mol Sci 2018; 19:ijms19071953. [PMID: 29973487 PMCID: PMC6073303 DOI: 10.3390/ijms19071953] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/28/2018] [Accepted: 06/30/2018] [Indexed: 12/11/2022] Open
Abstract
Macrophages, cells belonging to the innate immune system, present a high plasticity grade, being able to change their phenotype in response to environmental stimuli. They play central roles during development, homeostatic tissue processes, tissue repair, and immunity. Furthermore, it is recognized that macrophages are involved in chronic inflammation and that they play central roles in inflammatory diseases and cancer. Due to their large involvement in the pathogenesis of several types of human diseases, macrophages are considered to be relevant therapeutic targets. Nanotechnology-based systems have attracted a lot of attention in this field, gaining a pivotal role as useful moieties to target macrophages in diseased tissues. Among the different approaches that can target macrophages, the most radical is represented by their depletion, commonly obtained by means of clodronate-containing liposomal formulations and/or depleting antibodies. These strategies have produced encouraging results in experimental mouse models. In this review, we focus on macrophage targeting, based on the results so far obtained in preclinical models of inflammatory diseases and cancer. Pros and cons of these therapeutic interventions will be highlighted.
Collapse
|
429
|
Falconer J, Murphy AN, Young S, Clark AR, Tiziani S, Guma M, Buckley CD. Review: Synovial Cell Metabolism and Chronic Inflammation in Rheumatoid Arthritis. Arthritis Rheumatol 2018; 70:984-999. [PMID: 29579371 PMCID: PMC6019623 DOI: 10.1002/art.40504] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 03/15/2018] [Indexed: 12/17/2022]
Abstract
Metabolomic studies of body fluids show that immune-mediated inflammatory diseases such as rheumatoid arthritis (RA) are associated with metabolic disruption. This is likely to reflect the increased bioenergetic and biosynthetic demands of sustained inflammation and changes in nutrient and oxygen availability in damaged tissue. The synovial membrane lining layer is the principal site of inflammation in RA. Here, the resident cells are fibroblast-like synoviocytes (FLS) and synovial tissue macrophages, which are transformed toward overproduction of enzymes that degrade cartilage and bone and cytokines that promote immune cell infiltration. Recent studies have shown metabolic changes in both FLS and macrophages from RA patients, and these may be therapeutically targetable. However, because the origins and subset-specific functions of synoviocytes are poorly understood, and the signaling modules that control metabolic deviation in RA synovial cells are yet to be explored, significant additional research is needed to translate these findings to clinical application. Furthermore, in many inflamed tissues, different cell types can forge metabolic collaborations through solute carriers in their membranes to meet a high demand for energy or biomolecules. Such relationships are likely to exist in the synovium and have not been studied. Finally, it is not yet known whether metabolic change is a consequence of disease or whether primary changes to cellular metabolism might underlie or contribute to the pathogenesis of early-stage disease. In this review article, we collate what is known about metabolism in synovial tissue cells and highlight future directions of research in this area.
Collapse
Affiliation(s)
- Jane Falconer
- Rheumatology Research Group, Institute of inflammation and Ageing, College of Medical and dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Anne N Murphy
- Pharmacology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | - Stephen Young
- Rheumatology Research Group, Institute of inflammation and Ageing, College of Medical and dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Andrew R Clark
- Rheumatology Research Group, Institute of inflammation and Ageing, College of Medical and dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
| | - Stefano Tiziani
- Department of Nutritional Sciences & Dell Pediatric Research Institute, University of Texas at Austin, 1400 Barbara Jordan Blvd., Austin, TX
| | - Monica Guma
- Medicine, School of Medicine, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093
| | - Christopher D Buckley
- Rheumatology Research Group, Institute of inflammation and Ageing, College of Medical and dental Sciences, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford. UK
| |
Collapse
|
430
|
Bartlett DB, Willis LH, Slentz CA, Hoselton A, Kelly L, Huebner JL, Kraus VB, Moss J, Muehlbauer MJ, Spielmann G, Kraus WE, Lord JM, Huffman KM. Ten weeks of high-intensity interval walk training is associated with reduced disease activity and improved innate immune function in older adults with rheumatoid arthritis: a pilot study. Arthritis Res Ther 2018; 20:127. [PMID: 29898765 PMCID: PMC6001166 DOI: 10.1186/s13075-018-1624-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 05/14/2018] [Indexed: 12/15/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic inflammatory disease in which adults have significant joint issues leading to poor health. Poor health is compounded by many factors, including exercise avoidance and increased risk of opportunistic infection. Exercise training can improve the health of patients with RA and potentially improve immune function; however, information on the effects of high-intensity interval training (HIIT) in RA is limited. We sought to determine whether 10 weeks of a walking-based HIIT program would be associated with health improvements as measured by disease activity and aerobic fitness. Further, we assessed whether HIIT was associated with improved immune function, specifically antimicrobial/bacterial functions of neutrophils and monocytes. Methods Twelve physically inactive adults aged 64 ± 7 years with either seropositive or radiographically proven (bone erosions) RA completed 10 weeks of high-intensity interval walking. Training consisted of 3 × 30-minute sessions/week of ten ≥ 60-second intervals of high intensity (80–90% VO2reserve) separated by similar bouts of lower-intensity intervals (50–60% VO2reserve). Pre- and postintervention assessments included aerobic and physical function; disease activity as measured by Disease Activity score in 28 joints (DAS28), self-perceived health, C-reactive protein (CRP), and erythrocyte sedimentation rate (ESR); plasma interleukin (IL)-1β, IL-6, chemokine (C-X-C motif) ligand (CXCL)-8, IL-10, and tumor necrosis factor (TNF)-α concentrations; and neutrophil and monocyte phenotypes and functions. Results Despite minimal body composition change, cardiorespiratory fitness increased by 9% (change in both relative and absolute aerobic capacity; p < 0.001), and resting blood pressure and heart rate were both reduced (both p < 0.05). Postintervention disease activity was reduced by 38% (DAS28; p = 0.001) with significant reductions in ESR and swollen joints as well as improved self-perceived health. Neutrophil migration toward CXCL-8 (p = 0.003), phagocytosis of Escherichia coli (p = 0.03), and ROS production (p < 0.001) all increased following training. The frequency of cluster of differentiation 14-positive (CD14+)/CD16+ monocytes was reduced (p = 0.002), with both nonclassical (CD14dim/CD16bright) and intermediate (CD14bright/CD16positive) monocytes being reduced (both p < 0.05). Following training, the cell surface expression of intermediate monocyte Toll-like receptor 2 (TLR2), TLR4, and HLA-DR was reduced (all p < 0.05), and monocyte phagocytosis of E. coli increased (p = 0.02). No changes were observed for inflammatory markers IL-1β, IL-6, CXCL-8, IL-10, CRP, or TNF-α. Conclusions We report for the first time, to our knowledge, that a high-intensity interval walking protocol in older adults with stable RA is associated with reduced disease activity, improved cardiovascular fitness, and improved innate immune functions, indicative of reduced infection risk and inflammatory potential. Importantly, the exercise program was well tolerated by these patients. Trial registration ClinicalTrials.gov, NCT02528344. Registered on 19 August 2015.
Collapse
Affiliation(s)
- David B Bartlett
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA. .,Division of Medical Oncology, Duke University School of Medicine, Durham, NC, 27701, USA. .,MRC-ARUK Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.
| | - Leslie H Willis
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Cris A Slentz
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Andrew Hoselton
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Leslie Kelly
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Janet L Huebner
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Virginia B Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Jennifer Moss
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Michael J Muehlbauer
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | | | - William E Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| | - Janet M Lord
- MRC-ARUK Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.,NIHR Birmingham Biomedical Research Centre in Inflammation, University Hospital Birmingham, Birmingham, UK
| | - Kim M Huffman
- Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
431
|
Franko J, McCall JL, Barnett JB. Evaluating Macrophages in Immunotoxicity Testing. Methods Mol Biol 2018; 1803:255-296. [PMID: 29882145 DOI: 10.1007/978-1-4939-8549-4_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Macrophages are a heterogeneous group of cells that have a multitude of functions depending on their differentiation state. While classically known for their phagocytic and antigen presentation abilities, it is now evident that these cells fulfill homeostatic functions beyond the elimination of invading pathogens. In addition, macrophages have also been implicated in the downregulation of inflammatory responses following pathogen removal, tissue remodeling, repair, and angiogenesis. Alterations in macrophage differentiation and/or activity due to xenobiotic exposure can have grave consequences on organismal homeostasis, potentially contributing to disease due to immunosuppression or chronic inflammatory responses, depending upon the pathways affected. In this chapter, we provide an overview of the macrophages subtypes, their origin and a general discussion of several different assays used to assess their functional status.
Collapse
Affiliation(s)
- Jennifer Franko
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jamie L McCall
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - John B Barnett
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, USA.
| |
Collapse
|
432
|
Mandelin AM, Homan PJ, Shaffer AM, Cuda CM, Dominguez ST, Bacalao E, Carns M, Hinchcliff M, Lee J, Aren K, Thakrar A, Montgomery AB, Bridges SL, Bathon JM, Atkinson JP, Fox DA, Matteson EL, Buckley CD, Pitzalis C, Parks D, Hughes LB, Geraldino-Pardilla L, Ike R, Phillips K, Wright K, Filer A, Kelly S, Ruderman EM, Morgan V, Abdala-Valencia H, Misharin AV, Budinger GS, Bartom ET, Pope RM, Perlman H, Winter DR. Transcriptional Profiling of Synovial Macrophages Using Minimally Invasive Ultrasound-Guided Synovial Biopsies in Rheumatoid Arthritis. Arthritis Rheumatol 2018; 70:841-854. [PMID: 29439295 DOI: 10.1002/art.40453] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 02/08/2018] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Currently, there are no reliable biomarkers for predicting therapeutic response in patients with rheumatoid arthritis (RA). The synovium may unlock critical information for determining efficacy, since a reduction in the numbers of sublining synovial macrophages remains the most reproducible biomarker. Thus, a clinically actionable method for the collection of synovial tissue, which can be analyzed using high-throughput strategies, must become a reality. This study was undertaken to assess the feasibility of utilizing synovial biopsies as a precision medicine-based approach for patients with RA. METHODS Rheumatologists at 6 US academic sites were trained in minimally invasive ultrasound-guided synovial tissue biopsy. Biopsy specimens obtained from patients with RA and synovial tissue from patients with osteoarthritis (OA) were subjected to histologic analysis, fluorescence-activated cell sorting, and RNA sequencing (RNA-seq). An optimized protocol for digesting synovial tissue was developed to generate high-quality RNA-seq libraries from isolated macrophage populations. Associations were determined between macrophage transcriptional profiles and clinical parameters in RA patients. RESULTS Patients with RA reported minimal adverse effects in response to synovial biopsy. Comparable RNA quality was observed from synovial tissue and isolated macrophages between patients with RA and patients with OA. Whole tissue samples from patients with RA demonstrated a high degree of transcriptional heterogeneity. In contrast, the transcriptional profile of isolated RA synovial macrophages highlighted different subpopulations of patients and identified 6 novel transcriptional modules that were associated with disease activity and therapy. CONCLUSION Performance of synovial tissue biopsies by rheumatologists in the US is feasible and generates high-quality samples for research. Through the use of cutting-edge technologies to analyze synovial biopsy specimens in conjunction with corresponding clinical information, a precision medicine-based approach for patients with RA is attainable.
Collapse
Affiliation(s)
- Arthur M Mandelin
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Philip J Homan
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Carla M Cuda
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Emily Bacalao
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Mary Carns
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Jungwha Lee
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kathleen Aren
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Anjali Thakrar
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Anna B Montgomery
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | - John P Atkinson
- Washington University School of Medicine, St. Louis, Missouri
| | - David A Fox
- University of Michigan School of Medicine, Ann Arbor
| | - Eric L Matteson
- Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Christopher D Buckley
- University of Birmingham Research Laboratories, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Costantino Pitzalis
- William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Deborah Parks
- Washington University School of Medicine, St. Louis, Missouri
| | | | | | - Robert Ike
- University of Michigan School of Medicine, Ann Arbor
| | | | - Kerry Wright
- Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Andrew Filer
- University of Birmingham Research Laboratories, Queen Elizabeth Hospital Birmingham, Birmingham, UK
| | - Stephen Kelly
- William Harvey Research Institute and Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eric M Ruderman
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Vince Morgan
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | - G Scott Budinger
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Richard M Pope
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Harris Perlman
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Deborah R Winter
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
433
|
Zhong Q, Gong FY, Gong Z, Hua SH, Zeng KQ, Gao XM. IgG Immunocomplexes Sensitize Human Monocytes for Inflammatory Hyperactivity via Transcriptomic and Epigenetic Reprogramming in Rheumatoid Arthritis. THE JOURNAL OF IMMUNOLOGY 2018; 200:3913-3925. [PMID: 29712771 DOI: 10.4049/jimmunol.1701756] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 04/04/2018] [Indexed: 12/31/2022]
Abstract
Prevalence of circulating immunocomplexes (ICs) strongly correlates with rheumatoid arthritis (RA) in humans. Deposits of IgG-ICs are abundant in affected joints of patients, yet molecular mechanisms for the pathogenic roles of such ICs are not fully understood. In this study, we present evidence that IgG-ICs precipitated from RA sera sensitized human monocytes for a long-lasting inflammatory functional state, characterized by a strong TNF-α response to cellular proteins representing damage-associated molecular patterns and microbe-derived pathogen-associated molecular patterns. Importantly, plate-coated human IgG (a mimic of deposited IC without Ag restriction) exhibited a similarly robust ability of monocyte sensitization in vitro. The plate-coated human IgG-induced functional programming is accompanied by transcriptomic and epigenetic modification of various inflammatory cytokines and negative regulator genes. Moreover, macrophages freshly isolated from synovia of patients with RA, but not sera-negative arthropathy, displayed a signature gene expression profile highly similar to that of IC-sensitized human monocytes, indicative of historical priming events by IgG-ICs in vivo. Thus, the ability of IgG-ICs to drive sustainable functional sensitization/reprogramming of monocytes and macrophages toward inflammation may render them key players in the development of RA.
Collapse
Affiliation(s)
- Qiao Zhong
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China.,Department of Laboratory Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China.,Suzhou Municipal Hospital, Suzhou 215002, China
| | - Fang-Yuan Gong
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Zheng Gong
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Sheng-Hao Hua
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China
| | - Ke-Qin Zeng
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China.,Department of Rheumatology, The First Affiliated Hospital of Soochow University, Suzhou 215001, China
| | - Xiao-Ming Gao
- Institute of Biology and Medical Sciences, School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, China; .,Jiangsu Key Laboratory of Infection and Immunity, Suzhou 215123, China; and.,Key Laboratory of Systemic Biomedical Study, Suzhou 215123, China
| |
Collapse
|
434
|
Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT, Sahebkar A. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol 2018; 233:6425-6440. [PMID: 29319160 DOI: 10.1002/jcp.26429] [Citation(s) in RCA: 3096] [Impact Index Per Article: 442.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022]
Abstract
Macrophages are heterogeneous and their phenotype and functions are regulated by the surrounding micro-environment. Macrophages commonly exist in two distinct subsets: 1) Classically activated or M1 macrophages, which are pro-inflammatory and polarized by lipopolysaccharide (LPS) either alone or in association with Th1 cytokines such as IFN-γ, GM-CSF, and produce pro-inflammatory cytokines such as interleukin-1β (IL-1β), IL-6, IL-12, IL-23, and TNF-α; and 2) Alternatively activated or M2 macrophages, which are anti-inflammatory and immunoregulatory and polarized by Th2 cytokines such as IL-4 and IL-13 and produce anti-inflammatory cytokines such as IL-10 and TGF-β. M1 and M2 macrophages have different functions and transcriptional profiles. They have unique abilities by destroying pathogens or repair the inflammation-associated injury. It is known that M1/M2 macrophage balance polarization governs the fate of an organ in inflammation or injury. When the infection or inflammation is severe enough to affect an organ, macrophages first exhibit the M1 phenotype to release TNF-α, IL-1β, IL-12, and IL-23 against the stimulus. But, if M1 phase continues, it can cause tissue damage. Therefore, M2 macrophages secrete high amounts of IL-10 and TGF-β to suppress the inflammation, contribute to tissue repair, remodeling, vasculogenesis, and retain homeostasis. In this review, we first discuss the basic biology of macrophages including origin, differentiation and activation, tissue distribution, plasticity and polarization, migration, antigen presentation capacity, cytokine and chemokine production, metabolism, and involvement of microRNAs in macrophage polarization and function. Secondly, we discuss the protective and pathogenic role of the macrophage subsets in normal and pathological pregnancy, anti-microbial defense, anti-tumor immunity, metabolic disease and obesity, asthma and allergy, atherosclerosis, fibrosis, wound healing, and autoimmunity.
Collapse
Affiliation(s)
- Abbas Shapouri-Moghaddam
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Mohammadian
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Vazini
- Nursing Department, Basic Sciences Faculty, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Mahdi Taghadosi
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed-Alireza Esmaeili
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mardani
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bita Seifi
- Department of Anatomy, Islamic Azad University, Mashhad Branch, Iran
| | - Asadollah Mohammadi
- Inflammation and Inflammatory Disease Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil T Afshari
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
435
|
Kim B, Lee JH, Jin WJ, Kim HH, Ha H, Lee ZH. JN-2, a C-X-C motif chemokine receptor 3 antagonist, ameliorates arthritis progression in an animal model. Eur J Pharmacol 2018; 823:1-10. [DOI: 10.1016/j.ejphar.2018.01.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/18/2018] [Accepted: 01/23/2018] [Indexed: 12/11/2022]
|
436
|
Multiparametric Analysis of Myeloid Populations by Flow Cytometry. Methods Mol Biol 2018. [PMID: 29476466 DOI: 10.1007/978-1-4939-7680-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Flow cytometry is extensively used for the immune-profiling of leukocytes in tissue during homeostasis and inflammation. The multiparametric power of using fluorescently conjugated antibodies for specific surface and activation markers provides a comprehensive profile of immune cells. This chapter describes the identification and characterization of myeloid populations using flow cytometric analysis in an acute model of resolving inflammation. This model allows the examination of heterogenic populations across different systemic and tissue locations. We describe tissue processing, antibody staining, and analysis, which include a newly described viSNE tool to generate two-dimensional clustering within myeloid populations. We also reference the use of transgenic reporter mice on specific myeloid cells that provides enhanced specificity and profiling when defining myeloid heterogeneity.
Collapse
|
437
|
Imaging and Methotrexate Response Monitoring of Systemic Inflammation in Arthritic Rats Employing the Macrophage PET Tracer [ 18F]Fluoro-PEG-Folate. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:8092781. [PMID: 29681783 PMCID: PMC5841060 DOI: 10.1155/2018/8092781] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/22/2017] [Accepted: 01/24/2018] [Indexed: 12/23/2022]
Abstract
Background In rheumatoid arthritis, articular inflammation is a hallmark of disease, while the involvement of extra-articular tissues is less well defined. Here, we examined the feasibility of PET imaging with the macrophage tracer [18F]fluoro-PEG-folate, targeting folate receptor β (FRβ), to monitor systemic inflammatory disease in liver and spleen of arthritic rats before and after methotrexate (MTX) treatment. Methods [18F]Fluoro-PEG-folate PET scans (60 min) were acquired in saline- and MTX-treated (1 mg/kg, 4x) arthritic rats, followed by tissue resection and radiotracer distribution analysis. Liver and spleen tissues were stained for ED1/ED2-macrophage markers and FRβ expression. Results [18F]Fluoro-PEG-folate PET and ex vivo tissue distribution studies revealed a significant (p < 0.01) 2-fold lower tracer uptake in both liver and spleen of MTX-treated arthritic rats. Consistently, ED1- and ED2-positive macrophages were significantly (p < 0.01) decreased in liver (4-fold) and spleen (3-fold) of MTX-treated compared with saline-treated rats. Additionally, FRβ-positive macrophages were also significantly reduced in liver (5-fold, p < 0.005) and spleen (3-fold, p < 0.01) of MTX- versus saline-treated rats. Conclusions MTX treatment reduced activated macrophages in liver and spleen, as markers for systemic inflammation in these organs. Macrophage PET imaging with [18F]fluoro-PEG-folate holds promise for detection of systemic inflammation in RA as well as therapy (MTX) response monitoring.
Collapse
|
438
|
Silibinin alleviates inflammation and induces apoptosis in human rheumatoid arthritis fibroblast-like synoviocytes and has a therapeutic effect on arthritis in rats. Sci Rep 2018; 8:3241. [PMID: 29459717 PMCID: PMC5818498 DOI: 10.1038/s41598-018-21674-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 02/08/2018] [Indexed: 01/29/2023] Open
Abstract
Silibinin, a natural polyphenolic flavonoid, possesses anti-oxidant, anti-inflammation and anti-cancer properties. The present study was designed to investigate the effects of silibinin on rheumatoid arthritis (RA) pathogenesis-related cells and collagen-induced arthritis (CIA) and further explore the potential underlying mechanisms. Our results showed that silibinin suppressed cell viability and increased the percentage of apoptotic RA-fibroblast-like synoviocytes (FLS). Furthermore, the production of inflammatory cytokines in RA-FLS and a CIA rat model was effectively inhibited by silibinin. Silibinin also induced macrophage M2 polarization in RAW264.7 cells. We further demonstrated that silibinin inhibits Th17 cell differentiation in vitro. The nuclear factor kappa B (NF-κB) pathway was suppressed in RA-FLS. In addition, Sirtuin1 (SIRT1) was decreased after silibinin treatment, and RA-FLS transfection with a short hairpin RNA (shRNA) of SIRT1 enhanced silibinin-induced apoptosis. Autophagy was markedly decreased in a dose-dependent manner following silibinin treatment. These findings indicate that silibinin inhibited inflammation by inhibiting the NF-κB pathway, and SIRT1 may participate in silibinin-induced apoptosis. Silibinin also inhibited autophagy in RA-FLS. Thus, silibinin may be a potential therapeutic agent for the treatment of RA.
Collapse
|
439
|
Alivernini S, Tolusso B, Ferraccioli G, Gremese E, Kurowska-Stolarska M, McInnes IB. Driving chronicity in rheumatoid arthritis: perpetuating role of myeloid cells. Clin Exp Immunol 2018; 193:13-23. [PMID: 29315512 PMCID: PMC6038003 DOI: 10.1111/cei.13098] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 12/19/2017] [Accepted: 12/28/2017] [Indexed: 12/11/2022] Open
Abstract
Acute inflammation is a complex and tightly regulated homeostatic process that includes leucocyte migration from the vasculature into tissues to eliminate the pathogen/injury, followed by a pro‐resolving response promoting tissue repair. However, if inflammation is uncontrolled as in chronic diseases such as rheumatoid arthritis (RA), it leads to tissue damage and disability. Synovial tissue inflammation in RA patients is maintained by sustained activation of multiple inflammatory positive‐feedback regulatory pathways in a variety of cells, including myeloid cells. In this review, we will highlight recent evidence uncovering biological mechanisms contributing to the aberrant activation of myeloid cells that contributes to perpetuation of inflammation in RA, and discuss emerging data on anti‐inflammatory mediators contributing to sustained remission that may inform a novel category of therapeutic targets.
Collapse
Affiliation(s)
- S Alivernini
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - B Tolusso
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - G Ferraccioli
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - E Gremese
- Institute of Rheumatology, Fondazione Policlinico Universitario A. Gemelli - Catholic University of the Sacred Heart, Rome, Italy
| | - M Kurowska-Stolarska
- Institute of Infection, Immunity and Inflammation, University of Glasgow.,Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, UK
| | - I B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow.,Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, UK
| |
Collapse
|
440
|
Carini C, Hunter E, Ramadass AS, Green J, Akoulitchev A, McInnes IB, Goodyear CS. Chromosome conformation signatures define predictive markers of inadequate response to methotrexate in early rheumatoid arthritis. J Transl Med 2018; 16:18. [PMID: 29378619 PMCID: PMC5789697 DOI: 10.1186/s12967-018-1387-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/13/2018] [Indexed: 12/15/2022] Open
Abstract
Background There is a pressing need in rheumatoid arthritis (RA) to identify patients who will not respond to first-line disease-modifying anti-rheumatic drugs (DMARD). We explored whether differences in genomic architecture represented by a chromosome conformation signature (CCS) in blood taken from early RA patients before methotrexate (MTX) treatment could assist in identifying non-response to DMARD and, whether there is an association between such a signature and RA specific expression quantitative trait loci (eQTL). Methods We looked for the presence of a CCS in blood from early RA patients commencing MTX using chromosome conformation capture by EpiSwitch™. Using blood samples from MTX responders, non-responders and healthy controls, a custom designed biomarker discovery array was refined to a 5-marker CCS that could discriminate between responders and non-responders to MTX. We cross-validated the predictive power of the CCS by generating 150 randomized groups of 59 early RA patients (30 responders and 29 non-responders) before MTX treatment. The CCS was validated using a blinded, independent cohort of 19 early RA patients (9 responders and 10 non-responders). Last, the loci of the CCS markers were mapped to RA-specific eQTL. Results We identified a 5-marker CCS that could identify, at baseline, responders and non-responders to MTX. The CCS consisted of binary chromosome conformations in the genomic regions of IFNAR1, IL-21R, IL-23, CXCL13 and IL-17A. When tested on a cohort of 59 RA patients, the CCS provided a negative predictive value of 90.0% for MTX response. When tested on a blinded independent validation cohort of 19 early RA patients, the signature demonstrated a true negative response rate of 86 and a 90% sensitivity for detection of non-responders to MTX. Only conformations in responders mapped to RA-specific eQTL. Conclusions Here we demonstrate that detection of a CCS in blood in early RA is able to predict inadequate response to MTX with a high degree of accuracy. Our results provide a proof of principle that a priori stratification of response to MTX is possible, offering a mechanism to provide alternative treatments for non-responders to MTX earlier in the course of the disease. Electronic supplementary material The online version of this article (10.1186/s12967-018-1387-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claudio Carini
- Pfizer Inc., Cambridge, USA. .,Department of Asthma, Allergy & Lung Biology, GSTT Campus, King's College School of Medicine, London, UK.
| | | | | | | | | | | | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Carl S Goodyear
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
441
|
Gao CH, Dong HL, Tai L, Gao XM. Lactoferrin-Containing Immunocomplexes Drive the Conversion of Human Macrophages from M2- into M1-like Phenotype. Front Immunol 2018; 9:37. [PMID: 29410669 PMCID: PMC5787126 DOI: 10.3389/fimmu.2018.00037] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/05/2018] [Indexed: 12/29/2022] Open
Abstract
Macrophages are multifunctional cells that perform diverse roles in health and disease and considered the main source of inflammatory cytokines in affected joints of patients with rheumatoid arthritis (RA). M2 macrophages are well known as anti-inflammation and wound-healing cells; however, recent evidence suggests that they can also promote inflammation in RA, although the underlying mechanism remains to be clarified. Based upon our recent finding that lactoferrin (LTF)-containing IgG immunocomplex (LTF-IC), found elevated in RA sera, potent activators of human monocytes/macrophages, we herein demonstrate that LTF-IC was able to elicit immediate proinflammatory cytokine production by M2-polarized human macrophages through coligation with CD14/toll-like receptor (TLR) 4 and FcγRIIa (CD32a). The LTF-IC-treated M2 cells adopted surface maker expression profile similar to that of M1 phenotype and became functionally hyperactive to subsequent stimuli such as lipopolysaccharide, zymosan and IL-1β, which could provide a positive feedback signal to promote excessive inflammation in RA. They also acquired the ability to facilitate activation of Th17 cells that are known to play critical roles in RA pathology. We propose that IgG ICs containing TLR agonizing autoantigens are able to directly switch human macrophages from M2 into M1-like phenotype, thereby promoting excessive inflammation in autoimmune diseases such as RA.
Collapse
Affiliation(s)
- Chen-Hui Gao
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Hong-Liang Dong
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Li Tai
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Xiao-Ming Gao
- Institute of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
442
|
Almholt K, Hebsgaard JB, Nansen A, Andersson C, Pass J, Rønø B, Thygesen P, Pelzer H, Loftager M, Lund IK, Høyer-Hansen G, Frisch T, Jensen CH, Otte KS, Søe NH, Bartels EM, Andersen M, Bliddal H, Usher PA. Antibody-Mediated Neutralization of uPA Proteolytic Function Reduces Disease Progression in Mouse Arthritis Models. THE JOURNAL OF IMMUNOLOGY 2017; 200:957-965. [DOI: 10.4049/jimmunol.1701317] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 11/21/2017] [Indexed: 11/19/2022]
|
443
|
Kurowska-Stolarska M, Alivernini S. Synovial tissue macrophages: friend or foe? RMD Open 2017; 3:e000527. [PMID: 29299338 PMCID: PMC5729306 DOI: 10.1136/rmdopen-2017-000527] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/09/2017] [Accepted: 11/12/2017] [Indexed: 12/20/2022] Open
Abstract
Healthy synovial tissue includes a lining layer of synovial fibroblasts and macrophages. The influx of leucocytes during active rheumatoid arthritis (RA) includes monocytes that differentiate locally into proinflammatory macrophages, and these produce pathogenic tumour necrosis factor. During sustained remission, the synovial tissue macrophage numbers recede to normal. The constitutive presence of tissue macrophages in the lining layer of the synovial membrane in healthy donors and in patients with RA during remission suggests that this macrophage population may have a role in maintaining and reinstating synovial tissue homeostasis respectively. Recent appreciation of the different origins and functions of tissue-resident compared with monocyte-derived macrophages has improved the understanding of their relative involvement in organ homeostasis in mouse models of disease. In this review, informed by mouse models and human data, we describe the presence of different functional subpopulations of human synovial tissue macrophages and discuss their distinct contribution to joint homeostasis and chronic inflammation in RA.
Collapse
Affiliation(s)
- Mariola Kurowska-Stolarska
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.,Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Universities of Glasgow, Birmingham and Newcastle, Glasgow, Birmingham and Newcastle, UK
| | - Stefano Alivernini
- Institute of Rheumatology, Fondazione Policlinico Universitario A Gemelli, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
444
|
Abstract
Interferon (IFN)-γ can prime macrophages for inflammatory responses by several mechanisms, including enhancer establishment and gene activation. In this issue of Immunity, Kang et al. (2017) provide insight into the mechanisms of IFN-γ-mediated gene repression as they show that IFN-γ promotes the disassembly of select active enhancers by interfering with enhancer-binding transcription factor MAF.
Collapse
Affiliation(s)
- Boris Novakovic
- Department of Molecular Biology, Radboud Institute of Molecular Life Sciences and Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Cheng Wang
- Department of Molecular Biology, Radboud Institute of Molecular Life Sciences and Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands
| | - Colin Logie
- Department of Molecular Biology, Radboud Institute of Molecular Life Sciences and Faculty of Science, Radboud University, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
445
|
Wandering pathways in the regulation of innate immunity and inflammation. J Autoimmun 2017; 85:1-5. [PMID: 29079064 DOI: 10.1016/j.jaut.2017.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAM) have served as a paradigm of cancer-related inflammation. Moreover, investigations on TAM have led to the dissection of macrophage plasticity and polarization and to the discovery and analysis of molecular pathways of innate immunity, in particular cytokines, chemokines and PTX3 as a prototypic fluid phase pattern recognition molecule. Mechanisms of negative regulation are complex and include decoy receptors, receptor antagonists, anti-inflammatory cytokines and the signalling regulator IL-1R8. In this review, topics and open issues in relation to regulation of innate immunity and inflammation are discussed: 1) how macrophage and neutrophil plasticity and polarization underlie diverse pathological conditions ranging from autoimmunity to cancer and may pave the way to innovative diagnostic and therapeutic approaches; 2) the key role of decoy receptors and negative regulators (e.g. IL-1R2, ACKR2, IL-1R8) in striking a balance between amplification of immunity and resolution versus uncontrolled inflammation and tissue damage; 3) role of humoral innate immunity, illustrated by PTX3, in resistance against selected microbes, regulation of inflammation and immunity and tissue repair, with implications for diagnostic and therapeutic translation.
Collapse
|
446
|
The Role of High-Mobility Group Box-1 and Its Crosstalk with Microbiome in Rheumatoid Arthritis. Mediators Inflamm 2017; 2017:5230374. [PMID: 29200665 PMCID: PMC5672636 DOI: 10.1155/2017/5230374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/16/2017] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, definitely disabling, and potentially severe autoimmune disease. Although an increasing number of patients are affected, a key treatment for all patients has not been discovered. High-mobility group box-1 (HMGB1) is a nuclear protein passively and actively released by almost all cell types after several stimuli. HMGB1 is involved in RA pathogenesis, but a convincing explanation about its role and possible modulation in RA is still lacking. Microbiome and its homeostasis are altered in patients with RA, and the microbiota restoration has been proposed to patients with RA. The purpose of the present review is to analyze the available evidences regarding HMGB1 and microbiome roles in RA and the possible implications of the crosstalk between the nuclear protein and microbiome in understanding and possibly treating patients affected by this harmful condition.
Collapse
|
447
|
Saas P, Bonnefoy F, Toussirot E, Perruche S. Harnessing Apoptotic Cell Clearance to Treat Autoimmune Arthritis. Front Immunol 2017; 8:1191. [PMID: 29062314 PMCID: PMC5640883 DOI: 10.3389/fimmu.2017.01191] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/08/2017] [Indexed: 12/14/2022] Open
Abstract
Early-stage apoptotic cells possess immunomodulatory properties. Proper apoptotic cell clearance during homeostasis has been shown to limit subsequent immune responses. Based on these observations, early-stage apoptotic cell infusion has been used to prevent unwanted inflammatory responses in different experimental models of autoimmune diseases or transplantation. Moreover, this approach has been shown to be feasible without any toxicity in patients undergoing allogeneic hematopoietic cell transplantation to prevent graft-versus-host disease. However, whether early-stage apoptotic cell infusion can be used to treat ongoing inflammatory disorders has not been reported extensively. Recently, we have provided evidence that early-stage apoptotic cell infusion is able to control, at least transiently, ongoing collagen-induced arthritis. This beneficial therapeutic effect is associated with the modulation of antigen-presenting cell functions mainly of macrophages and plasmacytoid dendritic cells, as well as the induction of collagen-specific regulatory CD4+ T cells (Treg). Furthermore, the efficacy of this approach is not altered by the association with two standard treatments of rheumatoid arthritis (RA), methotrexate and tumor necrosis factor (TNF) inhibition. Here, in the light of these observations and recent data of the literature, we discuss the mechanisms of early-stage apoptotic cell infusion and how this therapeutic approach can be transposed to patients with RA.
Collapse
Affiliation(s)
- Philippe Saas
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Université Bourgogne Franche-Comté, Besançon, France.,INSERM CIC-1431, University Hospital of Besançon, Clinical Investigation Center in Biotherapy, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France
| | - Francis Bonnefoy
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Université Bourgogne Franche-Comté, Besançon, France
| | - Eric Toussirot
- INSERM CIC-1431, University Hospital of Besançon, Clinical Investigation Center in Biotherapy, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France.,Department of Rheumatology, University Hospital of Besançon, Besançon, France.,Department of Therapeutics, Université Bourgogne Franche-Comté, UPRES EA 4266, Pathogenic Agents and Inflammation, Besancon, France
| | - Sylvain Perruche
- INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Université Bourgogne Franche-Comté, Besançon, France
| |
Collapse
|
448
|
Srivastava S, Singh D, Patel S, Singh MR. Treatment of rheumatoid arthritis by targeting macrophages through folic acid tailored superoxide dismutase and serratiopeptidase. J Drug Deliv Sci Technol 2017. [DOI: 10.1016/j.jddst.2017.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
449
|
Lebrun A, Lo Re S, Chantry M, Izquierdo Carerra X, Uwambayinema F, Ricci D, Devosse R, Ibouraadaten S, Brombin L, Palmai-Pallag M, Yakoub Y, Pasparakis M, Lison D, Huaux F. CCR2 + monocytic myeloid-derived suppressor cells (M-MDSCs) inhibit collagen degradation and promote lung fibrosis by producing transforming growth factor-β1. J Pathol 2017; 243:320-330. [PMID: 28799208 DOI: 10.1002/path.4956] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 08/02/2017] [Accepted: 08/06/2017] [Indexed: 12/24/2022]
Abstract
Monocytes infiltrating scar tissue are predominantly viewed as progenitor cells. Here, we show that tissue CCR2+ monocytes have specific immunosuppressive and profibrotic functions. CCR2+ monocytic cells are acutely recruited to the lung before the onset of silica-induced fibrosis in mice. These tissue monocytes are defined as monocytic myeloid-derived suppressor cells (M-MDSCs) because they significantly suppress T-lymphocyte proliferation in vitro. M-MDSCs collected from silica-treated mice also express transforming growth factor (TGF)-β1, which stimulates lung fibroblasts to release tissue inhibitor of metalloproteinase (TIMP)-1, an inhibitor of metalloproteinase collagenolytic activity. By using LysMCreCCR2loxP/loxP mice, we show that limiting CCR2+ M-MDSC accumulation reduces the pulmonary contents of TGF-β1, TIMP-1 and collagen after silica treatment. M-MDSCs do not differentiate into lung macrophages, granulocytes or fibrocytes during pulmonary fibrogenesis. Collectively, our data indicate that M-MDSCs contribute to lung fibrosis by specifically promoting a non-degrading collagen microenvironment. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Astrid Lebrun
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Sandra Lo Re
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Mathilde Chantry
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Xavier Izquierdo Carerra
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Francine Uwambayinema
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Doriana Ricci
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Raynal Devosse
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Saloua Ibouraadaten
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Lisa Brombin
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Mihaly Palmai-Pallag
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Yousof Yakoub
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | | | - Dominique Lison
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - François Huaux
- Louvain Centre for Toxicology and Applied Pharmacology (LTAP), Institut de Recherche Experimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
450
|
Park SY, Lee SW, Lee SY, Hong KW, Bae SS, Kim K, Kim CD. SIRT1/Adenosine Monophosphate-Activated Protein Kinase α Signaling Enhances Macrophage Polarization to an Anti-inflammatory Phenotype in Rheumatoid Arthritis. Front Immunol 2017; 8:1135. [PMID: 28966618 PMCID: PMC5605563 DOI: 10.3389/fimmu.2017.01135] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/29/2017] [Indexed: 12/29/2022] Open
Abstract
Macrophages are crucially involved in the pathogenesis of rheumatoid arthritis (RA). Macrophages of the M1 phenotype act as pro-inflammatory mediators in synovium, whereas those of the M2 phenotype suppress inflammation and promote tissue repair. SIRT1 is a class 3 histone deacetylase with anti-inflammatory characteristics. However, the role played by SIRT1 in macrophage polarization has not been defined in RA. We investigated whether SIRT1 exerts anti-inflammatory effects by modulating M1/M2 polarization in macrophages from RA patients. In this study, SIRT1 activation promoted the phosphorylation of an adenosine monophosphate-activated protein kinase (AMPK) α/acetyl-CoA carboxylase in macrophages exposed to interleukin (IL)-4, and that this resulted in the expressions of M2 genes, including MDC, FcεRII, MrC1, and IL-10, at high levels. Furthermore, these expressions were inhibited by sirtinol (an inhibitor of SIRT1) and compound C (an inhibitor of AMPK). Moreover, SIRT1 activation downregulated LPS/interferon γ-mediated NF-κB activity by inhibiting p65 acetylation and the expression of M1 genes, such as CCL2, iNOS, IL-12 p35, and IL-12 p40. Macrophages from SIRT1 transgenic (Tg)-mice exhibited enhanced polarization of M2 phenotype macrophages and reduced polarization of M1 phenotype macrophages. In line with these observations, SIRT1-Tg mice showed less histological signs of arthritis, that is, lower TNFα and IL-1β expressions and less severe arthritis in the knee joints, compared to wild-type mice. Taken together, the study shows activation of SIRT1/AMPKα signaling exerts anti-inflammatory activities by regulating M1/M2 polarization, and thereby reduces inflammatory responses in RA. Furthermore, it suggests that SIRT1 signaling be viewed as a therapeutic target in RA.
Collapse
Affiliation(s)
- So Youn Park
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongsangnam-do, South Korea.,Gene and Cell Therapy Research Center for Vessel-Associated Diseases, Pusan National University, Gyeongsangnam-do, South Korea
| | - Sung Won Lee
- Department of Internal Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Sang Yeob Lee
- Department of Internal Medicine, College of Medicine, Dong-A University, Busan, South Korea
| | - Ki Whan Hong
- Gene and Cell Therapy Research Center for Vessel-Associated Diseases, Pusan National University, Gyeongsangnam-do, South Korea
| | - Sun Sik Bae
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongsangnam-do, South Korea.,Gene and Cell Therapy Research Center for Vessel-Associated Diseases, Pusan National University, Gyeongsangnam-do, South Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongsangnam-do, South Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Gyeongsangnam-do, South Korea.,Gene and Cell Therapy Research Center for Vessel-Associated Diseases, Pusan National University, Gyeongsangnam-do, South Korea
| |
Collapse
|