1
|
Jin Y, Xu C, Zhu Y, Gu Z. Extracellular vesicle as a next-generation drug delivery platform for rheumatoid arthritis therapy. J Control Release 2025; 381:113610. [PMID: 40058499 DOI: 10.1016/j.jconrel.2025.113610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disorder characterized by chronic inflammation and progressive damage to connective tissue. It is driven by dysregulated cellular homeostasis, often leading to autoimmune destruction and permanent disability in severe cases. Over the past decade, various drug delivery systems have been developed to enable targeted therapies for disease prevention, reduction, or suppression. As an emerging therapeutic platform, extracellular vesicles (EVs) offer several advantages over conventional drug delivery systems, including biocompatibility and low immunogenicity. Consequently, an increasing number of studies have explored EV-based delivery systems in the treatment of RA, leveraging their natural ability to evade phagocytosis, prolong in vivo half-life, and minimize the immunogenicity of therapeutic agents. In this review, we first provide an in-depth overview of the pathogenesis of RA and the current treatment landscape. We then discuss the classification and biological properties of EVs, their potential therapeutic mechanisms, and the latest advancements in EVs as drug delivery platforms for RA therapy. We emphasize the significance of EVs as carriers in RA treatment and their potential to revolutionize therapeutic strategies. Furthermore, we examine key technological innovations and the future trajectory of EV research, focusing on the challenges and opportunities in translating these platforms into clinical practice. Our discussion aims to offer a comprehensive understanding of the current state and future prospects of EV-based therapeutics in RA.
Collapse
Affiliation(s)
- Yi Jin
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States
| | - Yujuan Zhu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Zhifeng Gu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
2
|
Mishra B, Bachu M, Yuan R, Wingert C, Chaudhary V, Brauner C, Bell R, Ivashkiv LB. IL-10 targets IRF transcription factors to suppress IFN and inflammatory response genes by epigenetic mechanisms. Nat Immunol 2025; 26:748-759. [PMID: 40263613 DOI: 10.1038/s41590-025-02137-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 03/17/2025] [Indexed: 04/24/2025]
Abstract
Interleukin-10 (IL-10) is pivotal in suppressing innate immune activation, in large part by suppressing induction of inflammatory genes. Despite decades of research, the molecular mechanisms underlying this inhibition have not been resolved. Here we utilized an integrated epigenomic analysis to investigate IL-10-mediated suppression of LPS and TNF responses in primary human monocytes. Instead of inhibiting core TLR4-activated pathways such as NF-κB, MAPK-AP-1 and TBK1-IRF3 signaling, IL-10 targeted IRF transcription factor activity and DNA binding, particularly IRF5 and an IRF1-mediated amplification loop. This resulted in suppression of inflammatory NF-κB target genes and near-complete suppression of interferon-stimulated genes. Mechanisms of gene inhibition included downregulation of chromatin accessibility, de novo enhancer formation and IRF1-associated H3K27ac activating histone marks. These results provide a mechanism by which IL-10 suppresses inflammatory NF-κB target genes, highlight the role of IRF1 in inflammatory gene expression and describe the suppression of IFN responses by epigenetic mechanisms.
Collapse
Affiliation(s)
- Bikash Mishra
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Mahesh Bachu
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Ruoxi Yuan
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Claire Wingert
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Vidyanath Chaudhary
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Caroline Brauner
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Richard Bell
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Lionel B Ivashkiv
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA.
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Ohshima K, Torii S, Shimizu S. Presence of Gut Microbiota Worsens D-Galactosamine and Lipopolysaccharide-Induced Hepatic Injury in Mice. Genes Cells 2025; 30:e13183. [PMID: 39624985 DOI: 10.1111/gtc.13183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/19/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024]
Abstract
Acute liver failure is a serious, life-threatening disease. Although the gut microbiota has been considered to play a role in liver failure, the extent to which it is involved in the pathogenesis of this disease has not been fully elucidated to date. Therefore, we here analyzed the importance of the presence of intestinal microbiota in the pathogenesis of acute liver injury, using D-galactosamine (D-GalN)/lipopolysaccharide (LPS)-treated mice, which is a widely used experimental model of acute liver injury. First, administration of the antibiotic polymyxin B markedly alleviated liver injury. Liver injury was also reduced in germ-free mice, leading to the conclusion that the presence of intestinal microbiota aggravates D-GalN/LPS-induced liver injury. The amount of bacteria and LPS transferred from the gut to the blood was not increased by D-GalN/LPS, suggesting that the worsening of liver injury was not simply owing to the entry of bacteria into the circulation. In conclusion, acute liver injury in polymyxin B-pretreated or germ-free mice was ameliorated by modulation of the gut microbiota. Modification of the gut microbiota using polymyxin B may hence have the potential to alleviate acute liver injury in human patients.
Collapse
Affiliation(s)
- Kazuma Ohshima
- Department of Pathological Cell Biology, Advanced Research Initiative, Institute of Science Tokyo, Tokyo, Japan
| | - Satoru Torii
- Department of Pathological Cell Biology, Advanced Research Initiative, Institute of Science Tokyo, Tokyo, Japan
| | - Shigeomi Shimizu
- Department of Pathological Cell Biology, Advanced Research Initiative, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
4
|
Kim HY, Kang YJ, Kim DH, Jang J, Lee SJ, Kim G, Koh HB, Ko YE, Shin HM, Lee H, Yoo TH, Lee WW. Uremic toxin indoxyl sulfate induces trained immunity via the AhR-dependent arachidonic acid pathway in end-stage renal disease (ESRD). eLife 2024; 12:RP87316. [PMID: 38980302 PMCID: PMC11233136 DOI: 10.7554/elife.87316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024] Open
Abstract
Trained immunity is the long-term functional reprogramming of innate immune cells, which results in altered responses toward a secondary challenge. Despite indoxyl sulfate (IS) being a potent stimulus associated with chronic kidney disease (CKD)-related inflammation, its impact on trained immunity has not been explored. Here, we demonstrate that IS induces trained immunity in monocytes via epigenetic and metabolic reprogramming, resulting in augmented cytokine production. Mechanistically, the aryl hydrocarbon receptor (AhR) contributes to IS-trained immunity by enhancing the expression of arachidonic acid (AA) metabolism-related genes such as arachidonate 5-lipoxygenase (ALOX5) and ALOX5 activating protein (ALOX5AP). Inhibition of AhR during IS training suppresses the induction of IS-trained immunity. Monocytes from end-stage renal disease (ESRD) patients have increased ALOX5 expression and after 6 days training, they exhibit enhanced TNF-α and IL-6 production to lipopolysaccharide (LPS). Furthermore, healthy control-derived monocytes trained with uremic sera from ESRD patients exhibit increased production of TNF-α and IL-6. Consistently, IS-trained mice and their splenic myeloid cells had increased production of TNF-α after in vivo and ex vivo LPS stimulation compared to that of control mice. These results provide insight into the role of IS in the induction of trained immunity, which is critical during inflammatory immune responses in CKD patients.
Collapse
Affiliation(s)
- Hee Young Kim
- Department of Microbiology and Immunology, Seoul National University College of MedicineSeoulRepublic of Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Yeon Jun Kang
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, and BK21Plus Biomedical Science Project, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Dong Hyun Kim
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, and BK21Plus Biomedical Science Project, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Jiyeon Jang
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, and BK21Plus Biomedical Science Project, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Su Jeong Lee
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, and BK21Plus Biomedical Science Project, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Gwanghun Kim
- Department of Biomedical Sciences, College of Medicine and BK21Plus Biomedical Science Project, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Hee Byung Koh
- Department of Internal Medicine, College of Medicine, Yonsei UniversitySeoulRepublic of Korea
| | - Ye Eun Ko
- Department of Internal Medicine, College of Medicine, Yonsei UniversitySeoulRepublic of Korea
| | - Hyun Mu Shin
- Department of Biomedical Sciences, College of Medicine and BK21Plus Biomedical Science Project, Seoul National University College of MedicineSeoulRepublic of Korea
- Wide River Institute of Immunology, Seoul National UniversityHongcheonRepublic of Korea
| | - Hajeong Lee
- Division of Nephrology, Department of Internal Medicine, Seoul National University HospitalSeoulRepublic of Korea
| | - Tae-Hyun Yoo
- Division of Nephrology, Department of Internal Medicine, Yonsei University College of MedicineSeoulRepublic of Korea
| | - Won-Woo Lee
- Department of Microbiology and Immunology, Seoul National University College of MedicineSeoulRepublic of Korea
- Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul National University College of MedicineSeoulRepublic of Korea
- Laboratory of Autoimmunity and Inflammation (LAI), Department of Biomedical Sciences, and BK21Plus Biomedical Science Project, Seoul National University College of MedicineSeoulRepublic of Korea
- Seoul National University Cancer Research Institute; Ischemic/Hypoxic Disease Institute, Seoul National University Medical Research Center, Seoul National University Hospital Biomedical Research InstituteSeoulRepublic of Korea
| |
Collapse
|
5
|
Sikder S, Pierce D, Sarkar ER, McHugh C, Quinlan KGR, Giacomin P, Loukas A. Regulation of host metabolic health by parasitic helminths. Trends Parasitol 2024; 40:386-400. [PMID: 38609741 DOI: 10.1016/j.pt.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024]
Abstract
Obesity is a worldwide pandemic and major risk factor for the development of metabolic syndrome (MetS) and type 2 diabetes (T2D). T2D requires lifelong medical support to limit complications and is defined by impaired glucose tolerance, insulin resistance (IR), and chronic low-level systemic inflammation initiating from adipose tissue. The current preventative strategies include a healthy diet, controlled physical activity, and medication targeting hyperglycemia, with underexplored underlying inflammation. Studies suggest a protective role for helminth infection in the prevention of T2D. The mechanisms may involve induction of modified type 2 and regulatory immune responses that suppress inflammation and promote insulin sensitivity. In this review, the roles of helminths in counteracting MetS, and prospects for harnessing these protective mechanisms for the development of novel anti-diabetes drugs are discussed.
Collapse
Affiliation(s)
- Suchandan Sikder
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia.
| | - Doris Pierce
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia
| | - Eti R Sarkar
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Queensland 4878, Australia
| | - Connor McHugh
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns, Queensland 4878, Australia
| | - Kate G R Quinlan
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Paul Giacomin
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; Macrobiome Therapeutics Pty Ltd, Cairns, Queensland 4878, Australia
| | - Alex Loukas
- Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Queensland 4878, Australia; Macrobiome Therapeutics Pty Ltd, Cairns, Queensland 4878, Australia
| |
Collapse
|
6
|
Ranjitkar S, Krajewski D, Garcia C, Tedeschi C, Polukort SH, Rovatti J, Mire M, Blesso CN, Jellison E, Schneider SS, Ryan JJ, Mathias CB. IL-10 Differentially Promotes Mast Cell Responsiveness to IL-33, Resulting in Enhancement of Type 2 Inflammation and Suppression of Neutrophilia. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1407-1419. [PMID: 38497670 PMCID: PMC11018500 DOI: 10.4049/jimmunol.2300884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/26/2024] [Indexed: 03/19/2024]
Abstract
Mast cells (MCs) play critical roles in the establishment of allergic diseases. We recently demonstrated an unexpected, proinflammatory role for IL-10 in regulating MC responses. IL-10 enhanced MC activation and promoted IgE-dependent responses during food allergy. However, whether these effects extend to IgE-independent stimuli is not clear. In this article, we demonstrate that IL-10 plays a critical role in driving IL-33-mediated MC responses. IL-10 stimulation enhanced MC expansion and degranulation, ST2 expression, IL-13 production, and phospho-relA upregulation in IL-33-treated cells while suppressing TNF-α. These effects were partly dependent on endogenous IL-10 and further amplified in MCs coactivated with both IL-33 and IgE/Ag. IL-10's divergent effects also extended in vivo. In a MC-dependent model of IL-33-induced neutrophilia, IL-10 treatment enhanced MC responsiveness, leading to suppression of neutrophils and decreased TNF-α. In contrast, during IL-33-induced type 2 inflammation, IL-10 priming exacerbated MC activity, resulting in MC recruitment to various tissues, enhanced ST2 expression, induction of hypothermia, recruitment of eosinophils, and increased MCPT-1 and IL-13 levels. Our data elucidate an important role for IL-10 as an augmenter of IL-33-mediated MC responses, with implications during both allergic diseases and other MC-dependent disorders. IL-10 induction is routinely used as a prognostic marker of disease improvement. Our data suggest instead that IL-10 can enhance ST2 responsiveness in IL-33-activated MCs, with the potential to both aggravate or suppress disease severity depending on the inflammatory context.
Collapse
Affiliation(s)
- Saurav Ranjitkar
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269
| | - Dylan Krajewski
- Department of Pharmaceutical and Administrative Sciences, Western New England University, Springfield, MA 01119
| | - Chelsea Garcia
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269
| | - Caitlin Tedeschi
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269
| | - Stephanie H. Polukort
- Department of Pharmaceutical and Administrative Sciences, Western New England University, Springfield, MA 01119
| | - Jeffrey Rovatti
- Department of Pharmaceutical and Administrative Sciences, Western New England University, Springfield, MA 01119
| | - Mohamed Mire
- Department of Pharmaceutical and Administrative Sciences, Western New England University, Springfield, MA 01119
| | | | - Evan Jellison
- Department of Immunology, University of Connecticut, Farmington, CT 06030
| | - Sallie S. Schneider
- Pioneer Valley Life Sciences Institute, Baystate Medical Center, Springfield, MA 01199
| | - John J. Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284
| | - Clinton B. Mathias
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269
| |
Collapse
|
7
|
Bongartz H, Bradfield C, Gross J, Fraser I, Nita-Lazar A, Meier-Schellersheim M. IL-10 dependent adaptation allows macrophages to adjust inflammatory responses to TLR4 stimulation history. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587272. [PMID: 38654826 PMCID: PMC11037870 DOI: 10.1101/2024.03.28.587272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
During an infection, innate immune cells must adjust nature and strength of their responses to changing pathogen abundances. To determine how stimulation of the pathogen sensing TLR4 shapes subsequent macrophage responses, we systematically varied priming and restimulation concentrations of its ligand KLA. We find that different priming strengths have very distinct effects at multiple stages of the signaling response, including receptor internalization, MAPK activation, cytokine and chemokine production, and nuclear translocation and chromatin association of NFκB and IκB members. In particular, restimulation-induced TNF-α production required KLA doses equal to or greater than those used for prior exposure, indicating that macrophages can detect and adaptively respond to changing TLR4 stimuli. Interestingly, while such adaptation was dependent on the anti-inflammatory cytokine IL-10, exogenous concentrations of IL-10 corresponding to those secreted after strong priming did not exert suppressive effects on TNF-α without such prior priming, confirming the critical role of TLR4 stimulation history.
Collapse
Affiliation(s)
- H. Bongartz
- Computational Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - C. Bradfield
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - J. Gross
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - I.D.C. Fraser
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - A. Nita-Lazar
- Functional Cellular Networks Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - M. Meier-Schellersheim
- Computational Systems Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Babbe H, Sundberg TB, Tichenor M, Seierstad M, Bacani G, Berstler J, Chai W, Chang L, Chung DM, Coe K, Collins B, Finley M, Guletsky A, Lemke CT, Mak PA, Mathur A, Mercado-Marin EV, Metkar S, Raymond DD, Rives ML, Rizzolio M, Shaffer PL, Smith R, Smith J, Steele R, Steffens H, Suarez J, Tian G, Majewski N, Volak LP, Wei J, Desai PT, Ong LL, Koudriakova T, Goldberg SD, Hirst G, Kaushik VK, Ort T, Seth N, Graham DB, Plevy S, Venable JD, Xavier RJ, Towne JE. Identification of highly selective SIK1/2 inhibitors that modulate innate immune activation and suppress intestinal inflammation. Proc Natl Acad Sci U S A 2024; 121:e2307086120. [PMID: 38147543 PMCID: PMC10769863 DOI: 10.1073/pnas.2307086120] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 11/07/2023] [Indexed: 12/28/2023] Open
Abstract
The salt-inducible kinases (SIK) 1-3 are key regulators of pro- versus anti-inflammatory cytokine responses during innate immune activation. The lack of highly SIK-family or SIK isoform-selective inhibitors suitable for repeat, oral dosing has limited the study of the optimal SIK isoform selectivity profile for suppressing inflammation in vivo. To overcome this challenge, we devised a structure-based design strategy for developing potent SIK inhibitors that are highly selective against other kinases by engaging two differentiating features of the SIK catalytic site. This effort resulted in SIK1/2-selective probes that inhibit key intracellular proximal signaling events including reducing phosphorylation of the SIK substrate cAMP response element binding protein (CREB) regulated transcription coactivator 3 (CRTC3) as detected with an internally generated phospho-Ser329-CRTC3-specific antibody. These inhibitors also suppress production of pro-inflammatory cytokines while inducing anti-inflammatory interleukin-10 in activated human and murine myeloid cells and in mice following a lipopolysaccharide challenge. Oral dosing of these compounds ameliorates disease in a murine colitis model. These findings define an approach to generate highly selective SIK1/2 inhibitors and establish that targeting these isoforms may be a useful strategy to suppress pathological inflammation.
Collapse
Affiliation(s)
- Holger Babbe
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Thomas B. Sundberg
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Mark Tichenor
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Mark Seierstad
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Genesis Bacani
- Janssen Research and Development, LLC., San Diego, CA92121
| | - James Berstler
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Wenying Chai
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Leon Chang
- Janssen Research and Development, LLC., San Diego, CA92121
| | | | - Kevin Coe
- Janssen Research and Development, LLC., San Diego, CA92121
| | | | - Michael Finley
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Alexander Guletsky
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Christopher T. Lemke
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Puiying A. Mak
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Ashok Mathur
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | - Shailesh Metkar
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Donald D. Raymond
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | | | | | - Paul L. Shaffer
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Russell Smith
- Janssen Research and Development, LLC., San Diego, CA92121
| | | | - Ruth Steele
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | - Javier Suarez
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Gaochao Tian
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Nathan Majewski
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | - Jianmei Wei
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Prerak T. Desai
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Luvena L. Ong
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | | | - Gavin Hirst
- Janssen Research and Development, LLC., San Diego, CA92121
| | - Virendar K. Kaushik
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, Cambridge, MA02142
| | - Tatiana Ort
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Nilufer Seth
- Janssen Research and Development, LLC., Spring House, PA19477
| | - Daniel B. Graham
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA02114
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA02142
| | - Scott Plevy
- Janssen Research and Development, LLC., Spring House, PA19477
| | | | - Ramnik J. Xavier
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA02114
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA02142
| | | |
Collapse
|
9
|
Nikaein N, Tuerxun K, Cedersund G, Eklund D, Kruse R, Särndahl E, Nånberg E, Thonig A, Repsilber D, Persson A, Nyman E. Mathematical models disentangle the role of IL-10 feedbacks in human monocytes upon proinflammatory activation. J Biol Chem 2023; 299:105205. [PMID: 37660912 PMCID: PMC10556785 DOI: 10.1016/j.jbc.2023.105205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/16/2023] [Accepted: 08/24/2023] [Indexed: 09/05/2023] Open
Abstract
Inflammation is one of the vital mechanisms through which the immune system responds to harmful stimuli. During inflammation, proinflammatory and anti-inflammatory cytokines interplay to orchestrate fine-tuned and dynamic immune responses. The cytokine interplay governs switches in the inflammatory response and dictates the propagation and development of the inflammatory response. Molecular pathways underlying the interplay are complex, and time-resolved monitoring of mediators and cytokines is necessary as a basis to study them in detail. Our understanding can be advanced by mathematical models that enable to analyze the system of interactions and their dynamical interplay in detail. We, therefore, used a mathematical modeling approach to study the interplay between prominent proinflammatory and anti-inflammatory cytokines with a focus on tumor necrosis factor and interleukin 10 (IL-10) in lipopolysaccharide-primed primary human monocytes. Relevant time-resolved data were generated by experimentally adding or blocking IL-10 at different time points. The model was successfully trained and could predict independent validation data and was further used to perform simulations to disentangle the role of IL-10 feedbacks during an acute inflammatory event. We used the insight to obtain a reduced predictive model including only the necessary IL-10-mediated feedbacks. Finally, the validated reduced model was used to predict early IL-10-tumor necrosis factor switches in the inflammatory response. Overall, we gained detailed insights into fine-tuning of inflammatory responses in human monocytes and present a model for further use in studying the complex and dynamic process of cytokine-regulated acute inflammation.
Collapse
Affiliation(s)
- Niloofar Nikaein
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden.
| | - Kedeye Tuerxun
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Gunnar Cedersund
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden; Department of Biomedical Engineering, Linköping University, Linköping, Sweden; Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Daniel Eklund
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Robert Kruse
- Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Department of Clinical Research Laboratory, Örebro University, Örebro, Sweden
| | - Eva Särndahl
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Eewa Nånberg
- Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden; Faculty of Medicine and Health, School of Health Sciences, Örebro University, Örebro, Sweden
| | - Antje Thonig
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Dirk Repsilber
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Alexander Persson
- Faculty of Medicine and Health, School of Medical Sciences, Örebro University, Örebro, Sweden; Faculty of Medicine and Health, Inflammatory Response and Infection Susceptibility Centre (iRiSC), Örebro University, Örebro, Sweden
| | - Elin Nyman
- Department of Biomedical Engineering, Linköping University, Linköping, Sweden.
| |
Collapse
|
10
|
Kammala AK, Mosebarger A, Radnaa E, Rowlinson E, Vora N, Fortunato SJ, Sharma S, Safarzadeh M, Menon R. Extracellular Vesicles-mediated recombinant IL-10 protects against ascending infection-associated preterm birth by reducing fetal inflammatory response. Front Immunol 2023; 14:1196453. [PMID: 37600782 PMCID: PMC10437065 DOI: 10.3389/fimmu.2023.1196453] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Background Fetal inflammatory response mediated by the influx of immune cells and activation of pro-inflammatory transcription factor NF-κB in feto-maternal uterine tissues is the major determinant of infection-associated preterm birth (PTB, live births < 37 weeks of gestation). Objective To reduce the incidence of PTB by minimizing inflammation, extracellular vesicles (EVs) were electroporetically engineered to contain anti-inflammatory cytokine interleukin (IL)-10 (eIL-10), and their efficacy was tested in an ascending model of infection (vaginal administration of E. coli) induced PTB in mouse models. Study design EVs (size: 30-170 nm) derived from HEK293T cells were electroporated with recombinant IL-10 at 500 volts and 125 Ω, and 6 pulses to generate eIL-10. eIL-10 structural characters (electron microscopy, nanoparticle tracking analysis, ExoView [size and cargo content] and functional properties (co-treatment of macrophage cells with LPS and eIL-10) were assessed. To test efficacy, CD1 mice were vaginally inoculated with E. coli (1010CFU) and subsequently treated with either PBS, eIL-10 (500ng) or Gentamicin (10mg/kg) or a combination of eIL-10+gentamicin. Fetal inflammatory response in maternal and fetal tissues after the infection or treatment were conducted by suspension Cytometer Time of Flight (CyTOF) using a transgenic mouse model that express red fluorescent TdTomato (mT+) in fetal cells. Results Engineered EVs were structurally and functionally stable and showed reduced proinflammatory cytokine production from LPS challenged macrophage cells in vitro. Maternal administration of eIL-10 (10 µg/kg body weight) crossed feto-maternal barriers to delay E. coli-induced PTB to deliver live pups at term. Delay in PTB was associated with reduced feto-maternal uterine inflammation (immune cell infiltration and histologic chorioamnionitis, NF-κB activation, and proinflammatory cytokine production). Conclusions eIL-10 administration was safe, stable, specific, delayed PTB by over 72 hrs and delivered live pups. The delivery of drugs using EVs overcomes the limitations of in-utero fetal interventions. Protecting IL-10 in EVs eliminates the need for the amniotic administration of recombinant IL-10 for its efficacy.
Collapse
Affiliation(s)
- Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Angela Mosebarger
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Enkhtuya Radnaa
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Emma Rowlinson
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Natasha Vora
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Stephen J. Fortunato
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Surendra Sharma
- Department of Pediatrics, Women & Infants Hospital of Rhode Island, Providence, RI, United States
| | - Melody Safarzadeh
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
11
|
Chen N, Li M, Wu H, Qin Y, Wang J, Xu K, Luo R, Yang L, Wang Y, Zhang X. An extracellular matrix-mimetic coating with dual bionics for cardiovascular stents. Regen Biomater 2023; 10:rbad055. [PMID: 37359731 PMCID: PMC10287914 DOI: 10.1093/rb/rbad055] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Anti-inflammation and anti-coagulation are the primary requirements for cardiovascular stents and also the widely accepted trajectory for multi-functional modification. In this work, we proposed an extracellular matrix (ECM)-mimetic coating for cardiovascular stents with the amplified functionalization of recombinant humanized collagen type III (rhCOL III), where the biomimetics were driven by structure mimicry and component/function mimicry. Briefly, the structure-mimic was constructed by the formation of a nanofiber (NF) structure via the polymerization of polysiloxane with a further introduction of amine groups as the nanofibrous layer. The fiber network could function as a three-dimensional reservoir to support the amplified immobilization of rhCoL III. The rhCOL III was tailored for anti-coagulant, anti-inflammatory and endothelialization promotion properties, which endows the ECM-mimetic coating with desired surface functionalities. Stent implantation in the abdominal aorta of rabbits was conducted to validate the in vivo re-endothelialization of the ECM-mimetic coating. The mild inflammatory responses, anti-thrombotic property, promotion of endothelialization and suppression of excessive neointimal hyperplasia confirmed that the ECM-mimetic coating provided a promising approach for the modification of vascular implants.
Collapse
Affiliation(s)
- Nuoya Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Mingyu Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Haoshaung Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Yumei Qin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Jian Wang
- Shanxi Key Laboratory of Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd, Taiyuan 030032, Shanxi, China
| | - Kai Xu
- Department of Cardiology, General Hospital of Northern Theater Command, Shenyang 110000, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| | | | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
12
|
Lopes JA, Boeno CN, Paloschi MV, Silva MDS, Rego CMA, Pires WL, Santana HM, Chaves YO, Rodrigues MMDS, Lima AM, Setúbal SDS, Soares AM, Zuliani JP. Phenotypic, functional and plasticity features of human PBMCs induced by venom secreted PLA 2s. Mol Immunol 2023; 155:135-152. [PMID: 36812762 DOI: 10.1016/j.molimm.2023.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/22/2023]
Abstract
Bothrops venom contains a high amount of secreted phospholipase A2 (sPLA2s) enzymes responsible for the inflammatory reaction and activation of leukocytes in cases of envenoming. PLA2s are proteins that have enzymatic activity and can hydrolyze phospholipids at the sn-2 position, thereby releasing fatty acids and lysophospholipids precursors of eicosanoids, which are significant mediators of inflammatory conditions. Whether these enzymes have a role in the activation and function of peripheral blood mononuclear cells (PBMCs) is not known. Here we show for the first time how two secreted PLA2s (BthTX-I and BthTX-II) isolated from the venom of Bothrops jararacussu affect the function and polarization of PBMCs. Neither BthTX-I nor BthTX-II exhibited significant cytotoxicity to isolated PBMCs compared with the control at any of the time points studied. RT-qPCR and enzyme-linked immunosorbent assays were used to determine changes in gene expression and the release of pro-inflammatory (TNF-α, IL-6, and IL-12) and anti-inflammatory (TGF-β and IL-10) cytokines, respectively, during the cell differentiation process. Lipid droplets formation and phagocytosis were also investigated. Monocytes/macrophages were labeled with anti-CD14, -CD163, and -CD206 antibodies to assay cell polarization. Both toxins caused a heterogeneous morphology (M1 and M2) on days 1 and 7 based on immunofluorescence analysis, revealing the considerable flexibility of these cells even in the presence of typical polarization stimuli. Thus, these findings indicate that the two sPLA2s trigger both immune response profiles in PBMCs indicating a significant degree of cell plasticity, which may be crucial for understanding the consequences of snake envenoming.
Collapse
Affiliation(s)
- Jéssica Amaral Lopes
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Charles Nunes Boeno
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Mauro Valentino Paloschi
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Milena Daniela Souza Silva
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Cristina Matiele Alves Rego
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Weverson Luciano Pires
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Hallison Mota Santana
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Yury Oliveira Chaves
- Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Fundação Oswaldo Cruz, FIOCRUZ Amazônia, Manaus, AM, Brazil
| | - Moreno Magalhães de Souza Rodrigues
- Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Laboratório de Análise e Visualização de Dados, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil
| | - Anderson M Lima
- Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Brazil
| | - Sulamita da S Setúbal
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil
| | - Andreimar M Soares
- Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Brazil
| | - Juliana P Zuliani
- Laboratório de Imunologia Celular Aplicada à Saúde, Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Porto Velho, RO, Brazil; Departamento de Medicina, Universidade Federal de Rondônia, UNIR, Porto Velho, RO, Brazil; Laboratório de Biotecnologia de Proteínas e Compostos Bioativos, LABIOPROT, Centro de Estudos de Biomoléculas Aplicadas à Saúde (CEBio), Fundação Oswaldo Cruz, FIOCRUZ Rondônia, Brazil.
| |
Collapse
|
13
|
Della Guardia L, Codella R. Exercise Restores Hypothalamic Health in Obesity by Reshaping the Inflammatory Network. Antioxidants (Basel) 2023; 12:antiox12020297. [PMID: 36829858 PMCID: PMC9951965 DOI: 10.3390/antiox12020297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Obesity and overnutrition induce inflammation, leptin-, and insulin resistance in the hypothalamus. The mediobasal hypothalamus responds to exercise enabling critical adaptions at molecular and cellular level that positively impact local inflammation. This review discusses the positive effect of exercise on obesity-induced hypothalamic dysfunction, highlighting the mechanistic aspects related to the anti-inflammatory effects of exercise. In HFD-fed animals, both acute and chronic moderate-intensity exercise mitigate microgliosis and lower inflammation in the arcuate nucleus (ARC). Notably, this associates with restored leptin sensitivity and lower food intake. Exercise-induced cytokines IL-6 and IL-10 mediate part of these positive effect on the ARC in obese animals. The reduction of obesity-associated pro-inflammatory mediators (e.g., FFAs, TNFα, resistin, and AGEs), and the improvement in the gut-brain axis represent alternative paths through which regular exercise can mitigate hypothalamic inflammation. These findings suggest that the regular practice of exercise can restore a proper functionality in the hypothalamus in obesity. Further analysis investigating the crosstalk muscle-hypothalamus would help toward a deeper comprehension of the subject.
Collapse
Affiliation(s)
- Lucio Della Guardia
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy
| | - Roberto Codella
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy
- Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, 20138 Milan, Italy
- Correspondence: ; Tel.: +39-02-50330356
| |
Collapse
|
14
|
Pei X, Heng X, Chu W. Polystyrene nano/microplastics induce microbiota dysbiosis, oxidative damage, and innate immune disruption in zebrafish. Microb Pathog 2022; 163:105387. [PMID: 34990781 DOI: 10.1016/j.micpath.2021.105387] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 11/29/2022]
Abstract
The toxicity of polystyrene nano/microplastics with diameter sizes of 50um and 100 nm and concentrations of 100 and 1000 μg/mL on gut microbiota, antioxidant activity and innate immune response in zebrafish was investigated. After exposure to polystyrene plastics particle, the pathological morphological changes of intestine and gills were observed, and the injury severity was related to the concentration and particle size of plastics. Significant changes in the richness and diversity of gut microbiota were observed after polystyrene plastics-exposed in zebrafish. The plastics-treated groups exhibited more substantial oxidative stress than the control group. In addition, the mRNA expression level of most pro- and anti-inflammatory factors, including IL-8, NF-κb, and IL-10, increased while the mRNA expression of TNF-α, a pro-inflammatory factor, decreased. Our results suggest that polystyrene nano/microplastics may represent a potential threat to the gut microbiota, oxidative status, and innate immunity. These results indicated that polystyrene nano/microplastics exerted size and concentration-dependent toxicity on zebrafish. The findings provide new evidence for the toxicity of polystyrene plastics on zebrafish.
Collapse
Affiliation(s)
- Xin Pei
- Department of Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Xing Heng
- Department of Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Weihua Chu
- Department of Microbiology, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
15
|
Poledne R, Kralova Lesna I. Adipose tissue macrophages and atherogenesis – a synergy with cholesterolaemia. Physiol Res 2021. [DOI: 10.33549//physiolres.934745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Excessive LDL cholesterol concentration together with subclinical inflammation, in which macrophages play a central role, are linked pathologies. The process starts with the accumulation of macrophages in white adipose tissue and the switch of their polarization toward a pro-inflammatory phenotype. The proportion of pro-inflammatory macrophages in adipose tissue is related to the main risk predictors of cardiovascular disease. The cholesterol content of phospholipids of cell membranes seems to possess a crucial role in the regulation of membrane signal transduction and macrophage polarization. Also, different fatty acids of membrane phospholipids influence phenotypes of adipose tissue macrophages with saturated fatty acids stimulating pro-inflammatory whereas ω3 fatty acids anti-inflammatory changes. The inflammatory status of white adipose tissue, therefore, reflects not only adipose tissue volume but also adipose tissue macrophages feature. The beneficial dietary change leading to an atherogenic lipoprotein decrease may therefore synergically reduce adipose tissue driven inflammation.
Collapse
Affiliation(s)
- R Poledne
- Laboratory for Atherosclerosis Research, Institute for Clinical and Experimental Medicine, Prague, Czech Republic.
| | | |
Collapse
|
16
|
Della Guardia L, Codella R. Exercise tolls the bell for key mediators of low-grade inflammation in dysmetabolic conditions. Cytokine Growth Factor Rev 2021; 62:83-93. [PMID: 34620559 DOI: 10.1016/j.cytogfr.2021.09.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/30/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022]
Abstract
Metabolic conditions share a common low-grade inflammatory milieu, which represents a key-factor for their ignition and maintenance. Exercise is instrumental for warranting systemic cardio-metabolic balance, owing to its regulatory effect on inflammation. This review explores the effect of physical activity in the modulation of sub-inflammatory framework characterizing dysmetabolic conditions. Regular exercise suppresses plasma levels of TNFα, IL-1β, FFAs and MCP-1, in dysmetabolic subjects. In addition, a single session of training increases the anti-inflammatory IL-10, IL-1 receptor antagonist (IL-1ra), and muscle-derived IL-6, mitigating low-grade inflammation. Resting IL-6 levels are decreased in trained-dysmetabolic subjects, compared to sedentary. On the other hand, the acute release of muscle-IL-6, after exercise, seems to exert a regulatory effect on the metabolic and inflammatory balance. In fact, muscle-released IL-6 is presumably implicated in fat loss and boosts plasma levels of IL-10 and IL-1ra. The improvement of adipose tissue functionality, following regular exercise, is also critical for the mitigation of sub-inflammation. This effect is likely mediated by muscle-released IL-15 and IL-6 and partly relies on the brown-shifting of white adipocytes, induced by exercise. In obese-dysmetabolic subjects, moderate training is shown to restore gut-microbiota health, and this mitigates the translocation of bacterial-LPS into bloodstream. Finally, regular exercise can lower plasma advanced glycated endproducts. The articulated physiology of circulating mediators and the modulating effect of the pathophysiological background, render the comprehension of the exercise-regulatory effect on sub-inflammation a key issue, in dysmetabolism.
Collapse
Affiliation(s)
- Lucio Della Guardia
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Fratelli Cervi 93, Segrate, 20090 Milano, Italy
| | - Roberto Codella
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Via Fratelli Cervi 93, Segrate, 20090 Milano, Italy; Department of Endocrinology, Nutrition and Metabolic Diseases, IRCCS MultiMedica, Milano, Italy.
| |
Collapse
|
17
|
Regulation of the Nfkbiz Gene and Its Protein Product IkBζ in Animal Models of Sepsis and Endotoxic Shock. Infect Immun 2021; 89:IAI.00674-20. [PMID: 33431705 DOI: 10.1128/iai.00674-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
Sepsis is a life-threatening condition that arises from a poorly regulated inflammatory response to pathogenic organisms. Current treatments are limited to antibiotics, fluid resuscitation, and other supportive therapies. New targets for monitoring disease progression and therapeutic interventions are therefore critically needed. We previously reported that lipocalin-2 (Lcn2), a bacteriostatic mediator with potent proapoptotic activities, was robustly induced in sepsis. Other studies showed that Lcn2 was a predictor of mortality in septic patients. However, how Lcn2 is regulated during sepsis is poorly understood. We evaluated how IkBζ, an inducer of Lcn2, was regulated in sepsis using both the cecal ligation and puncture (CLP) and endotoxemia (lipopolysaccharide [LPS]) animal models. We show that Nfkbiz, the gene encoding IkBζ, was rapidly stimulated but, unlike Lcn2, whose expression persists during sepsis, mRNA levels of Nfkbiz decline to near basal levels several hours after its induction. In contrast, we observed that IkBζ expression remained highly elevated in septic animals following CLP but not LPS, indicating the occurrence of a CLP-specific mechanism that extends IkBζ half-life. By using an inhibitor of IkBζ, we determined that the expression of Lcn2 was largely controlled by IkBζ. Altogether, these data indicate that the high IkBζ expression in tissues likely contributes to the elevated expression of Lcn2 in sepsis. Since IkBζ is also capable of promoting or repressing other inflammatory genes, it might exert a central role in sepsis.
Collapse
|
18
|
Qian S, Tang Y, Tang QQ. Adipose tissue plasticity and the pleiotropic roles of BMP signaling. J Biol Chem 2021; 296:100678. [PMID: 33872596 PMCID: PMC8131923 DOI: 10.1016/j.jbc.2021.100678] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 04/11/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Adipose tissues, including white, beige, and brown adipose tissue, have evolved to be highly dynamic organs. Adipose tissues undergo profound changes during development and regeneration and readily undergo remodeling to meet the demands of an everchanging metabolic landscape. The dynamics are determined by the high plasticity of adipose tissues, which contain various cell types: adipocytes, immune cells, endothelial cells, nerves, and fibroblasts. There are numerous proteins that participate in regulating the plasticity of adipose tissues. Among these, bone morphogenetic proteins (BMPs) were initially found to regulate the differentiation of adipocytes, and they are being reported to have pleiotropic functions by emerging studies. Here, in the first half of the article, we summarize the plasticity of adipocytes and macrophages, which are two groups of cells targeted by BMP signaling in adipose tissues. We then review how BMPs regulate the differentiation, death, and lipid metabolism of adipocytes. In addition, the potential role of BMPs in regulating adipose tissue macrophages is considered. Finally, the expression of BMPs in adipose tissues and their metabolic relevance are discussed.
Collapse
Affiliation(s)
- Shuwen Qian
- The Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Tang
- The Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi-Qun Tang
- The Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, and Department of Endocrinology and Metabolism of Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Vitamin D as a Key Player in Modulating Rheumatoid Arthritis-derived Immune Response. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2020. [DOI: 10.22207/jpam.14.4.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic inflammatory disease with chronic nature of joints related to autoimmunity. Vitamin D was found to modulate cell growth, function of immune cells and anti-inflammatory action. The aims of that study were to investigate serum level of vitamin D and some cytokines and to identify the correlation between vitamin D and these cytokines in RA. Totally 40 RA patients without vitamin D supplement were involved in this study. Serum level of vitamin D, interleukin-6 (IL-6), IL-10, IL-35, C-reactive protein (CRP) and tumor necrosis factor α (TNF-α), all of them were measure in all patients by enzyme-linked immunosorbent assay (ELISA). Patients were classified according to Vitamin D levels into two groups; RA patients with Vit. D deficiency (n=25) and RA patients with Vit. D sufficiency (n=15). IL-6 was lower significantly (P = 0.03) in RA patients with Vit. D sufficiency than RA patients with Vit. D deficiency. IL-10 and IL-35 were higher significantly (P = 0.0234, P = 0.0356 respectively) in RA patients with Vit. D sufficiency than RA patients with Vit. D deficiency. Vit. D was significantly positively correlated with both IL-10 (r = 0.4516, P = 0.0034) and IL-35 (r = 0.3424, P = 0.0329) and negatively correlated with IL-6 (r = -0.3188, P = 0.0479). Sufficient serum level of Vit. D is correlated with higher level of anti-inflammatory cytokines (IL-10 and IL-35) and lower level of IL-6. This support the immunomodulatory effect of Vit. D in RA.
Collapse
|
20
|
Suzuki H, Yamazaki T, Ohshio K, Sugamata M, Yoshikawa M, Kanauchi O, Morita Y. A Specific Strain of Lactic Acid Bacteria, Lactobacillus paracasei, Inhibits Inflammasome Activation In Vitro and Prevents Inflammation-Related Disorders. THE JOURNAL OF IMMUNOLOGY 2020; 205:811-821. [PMID: 32591398 DOI: 10.4049/jimmunol.1900657] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 06/04/2020] [Indexed: 12/23/2022]
Abstract
Some strains of lactic acid bacteria (LAB) have anti-inflammatory effects, but the mechanism underlying the alleviation of inflammation by LAB is not fully understood. In this study, we examined the inhibitory effect of a certain strain of LAB, Lactobacillus paracasei, on inflammasome activation, which is associated with various inflammatory disorders. Using bone marrow-derived macrophages from BALB/c mice, we found that L. paracasei, but not L. rhamnosus, suppressed NLRP3 inflammasome activation and inhibited subsequent caspase-1 activation and IL-1β secretion. L. paracasei also had inhibitory effects on AIM2 and NLRC4 inflammasome activation as well as the NLRP3 inflammasome. These inhibitory effects of L. paracasei on inflammasome activation were dependent on autocrine IL-10 induced by L. paracasei-stimulated macrophages. Furthermore, IL-10 production by L. paracasei-stimulated macrophages was involved with phagocytosis and the NOD2 signaling pathway in macrophages. In addition to in vitro studies, oral administration of L. paracasei in C57BL/6 mice reduced monosodium urate crystal-induced peritoneal inflammation in vivo. Moreover, continuous intake of L. paracasei in C57BL/6 mice alleviated high fat diet-induced insulin resistance and aging-induced expression of biomarkers for T cell senescence. Taken together, we demonstrated that L. paracasei inhibits inflammasome activation in vitro and exhibits an anti-inflammatory function in vivo. These results indicate that LAB that have inhibitory effects on inflammasome activation might contribute to the alleviation of inflammation-related disorders.
Collapse
Affiliation(s)
- Hiroaki Suzuki
- Research Laboratories for Health Science and Food Technologies, Kirin Holdings Company, Tokyo 236-0004, Japan
| | - Takahiro Yamazaki
- Research Laboratories for Health Science and Food Technologies, Kirin Holdings Company, Tokyo 236-0004, Japan
| | - Konomi Ohshio
- Research Laboratories for Health Science and Food Technologies, Kirin Holdings Company, Tokyo 236-0004, Japan
| | - Miho Sugamata
- Research Laboratories for Health Science and Food Technologies, Kirin Holdings Company, Tokyo 236-0004, Japan
| | - Mia Yoshikawa
- Research Laboratories for Health Science and Food Technologies, Kirin Holdings Company, Tokyo 236-0004, Japan
| | - Osamu Kanauchi
- Research Laboratories for Health Science and Food Technologies, Kirin Holdings Company, Tokyo 236-0004, Japan
| | - Yuji Morita
- Research Laboratories for Health Science and Food Technologies, Kirin Holdings Company, Tokyo 236-0004, Japan
| |
Collapse
|
21
|
Ho KV, Schreiber KL, Park J, Vo PH, Lei Z, Sumner LW, Brown CR, Lin CH. Identification and Quantification of Bioactive Molecules Inhibiting Pro-inflammatory Cytokine Production in Spent Coffee Grounds Using Metabolomics Analyses. Front Pharmacol 2020; 11:229. [PMID: 32210815 PMCID: PMC7073796 DOI: 10.3389/fphar.2020.00229] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/19/2020] [Indexed: 11/29/2022] Open
Abstract
In this study, we assessed the anti-inflammatory properties of spent coffee grounds. Methanolic extracts of spent coffee grounds obtained from 3 Arabica cultivars possess compounds that exerted inhibitory effects on the secretion of inflammatory mediators (TNF-α, IL-6, and IL-10) induced by a human pro-monocytic cell line differentiated with PMA and stimulated with lipopolysaccharide (LPS). Our results indicated that the cytokine suppressive activities of the spent coffee ground (SCG) extracts were different among coffee cultivars tested. Hawaiian Kona extracts exhibited inhibitory effects on the expression of 3 examined cytokines, Ethiopian Yirgacheffe extracts reduced the secretion of TNF-α and IL-6, and Costa Rican Tarrazu extracts decreased the secretion of IL-6 only. Untargeted metabolomics analyses of SCG extracts led to the putative identification of 26 metabolites with known anti-inflammatory activities. Multiple metabolites (i.e., chrysin, daidzein, eugenol, naringenin, naringin, oxyresveratrol, pectolinarin, resveratrol, tectochrysin, theaflavin, vanillic acid, and vitexin rhamnoside) identified in the SCGs represent possible novel anti-inflammatory compounds. Of the 26 identified metabolites, the 12 compounds that had high relative intensities in all of the extracts were successfully quantified using liquid chromatography-tandem mass spectrometry analyses. Results from the targeted analyses indicated that caffeine and 5-caffeoylquinic acid (CQA) were the most abundant compounds in the SCG extracts. The contents of caffeine ranged from 0.38 mg/g (Ethiopian Yirgacheffe) – 0.44 mg/g (Costa Rican Tarrazu), whereas 5-CQA concentrations were in the range of 0.24 mg/g (Costa Rican Tarrazu) – 0.34 mg/g (Ethiopian Yirgacheffe). The presence of multiple anti-inflammatory compounds in SCGs provides a promising natural source for cosmetic and pharmaceutical industries.
Collapse
Affiliation(s)
- Khanh-Van Ho
- Center for Agroforestry, School of Natural Resources, University of Missouri, Columbia, MO, United States.,Department of Food Technology, Can Tho University, Can Tho, Vietnam
| | - Kathy L Schreiber
- Cell and Immunobiology Core, University of Missouri, Columbia, MO, United States
| | - Jihyun Park
- Center for Agroforestry, School of Natural Resources, University of Missouri, Columbia, MO, United States
| | - Phuc H Vo
- Center for Agroforestry, School of Natural Resources, University of Missouri, Columbia, MO, United States
| | - Zhentian Lei
- Metabolomics Center, University of Missouri, Columbia, MO, United States.,Department of Biochemistry, Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Lloyd W Sumner
- Metabolomics Center, University of Missouri, Columbia, MO, United States.,Department of Biochemistry, Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Charles R Brown
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Chung-Ho Lin
- Center for Agroforestry, School of Natural Resources, University of Missouri, Columbia, MO, United States
| |
Collapse
|
22
|
Chen Z, Bozec A, Ramming A, Schett G. Anti-inflammatory and immune-regulatory cytokines in rheumatoid arthritis. Nat Rev Rheumatol 2020; 15:9-17. [PMID: 30341437 DOI: 10.1038/s41584-018-0109-2] [Citation(s) in RCA: 462] [Impact Index Per Article: 92.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by a failure of spontaneous resolution of inflammation. Although the pro-inflammatory cytokines and mediators that trigger RA have been the focus of intense investigations, the regulatory and anti-inflammatory cytokines responsible for the suppression and resolution of disease in a context-dependent manner have been less well characterized. However, knowledge of the pathways that control the suppression and resolution of inflammation in RA is clinically relevant and conceptually important for understanding the pathophysiology of the disease and for the development of treatments that enable long-term remission. Cytokine-mediated processes such as the activation of T helper 2 cells by IL-4 and IL-13, the resolution of inflammation by IL-9, IL-5-induced eosinophil expansion, IL-33-mediated macrophage polarization, the production of IL-10 by regulatory B cells and IL-27-mediated suppression of lymphoid follicle formation are all involved in governing the regulation and resolution of inflammation in RA. By better understanding these immune-regulatory signalling pathways, new therapeutic strategies for RA can be envisioned that aim to balance and resolve, rather than suppress, inflammation.
Collapse
Affiliation(s)
- Zhu Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of the University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Aline Bozec
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum Erlangen, Erlangen, Germany
| | - Andreas Ramming
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitatsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
23
|
Vega-Galaviz D, Vecchyo-Tenorio GD, Alcántara-Suárez R, Méndez-García LA, Sánchez-Del Real AL, Villalobos-Molina R, Fragoso JM, León-Cabrera S, Ostoa-Saloma P, Pérez-Tamayo R, Escobedo G. M2 macrophage immunotherapy abolishes glucose intolerance by increasing IL-10 expression and AKT activation. Immunotherapy 2020; 12:9-24. [PMID: 31914828 DOI: 10.2217/imt-2019-0080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aim: Glucose intolerance associates with M1/M2 macrophage unbalance. We thus wanted to examine the effect of M2 macrophage administration on mouse model of glucose intolerance. Materials & methods: C57BL/6 mice fed a high-fat diet (HFD) for 12 weeks and then received thrice 20 mg/kg streptozotocin (HFD-GI). Bone marrow-derived stem cells were collected from donor mice and differentiated/activated into M2 macrophages for intraperitoneal administration into HFD-GI mice. Results: M2 macrophage treatment abolished glucose intolerance independently of obesity. M2 macrophage administration increased IL-10 in visceral adipose tissue and serum, but showed no effect on serum insulin. While nitric oxide synthase-2 and arginase-1 remained unaltered, M2 macrophage treatment restored AKT phosphorylation in visceral adipose tissue. Conclusion: M2 macrophage treatment abolishes glucose intolerance by increasing IL-10 and phosphorylated AKT.
Collapse
Affiliation(s)
- Diana Vega-Galaviz
- Laboratory for Proteomics & Metabolomics, Research Division, General Hospital of Mexico 'Dr. Eduardo Liceaga', 06720 Mexico City, Mexico
| | - Georgina Del Vecchyo-Tenorio
- Laboratory for Proteomics & Metabolomics, Research Division, General Hospital of Mexico 'Dr. Eduardo Liceaga', 06720 Mexico City, Mexico.,Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Raúl Alcántara-Suárez
- Laboratory for Proteomics & Metabolomics, Research Division, General Hospital of Mexico 'Dr. Eduardo Liceaga', 06720 Mexico City, Mexico
| | - Lucia A Méndez-García
- Laboratory for Proteomics & Metabolomics, Research Division, General Hospital of Mexico 'Dr. Eduardo Liceaga', 06720 Mexico City, Mexico
| | - Ana L Sánchez-Del Real
- Laboratory for Proteomics & Metabolomics, Research Division, General Hospital of Mexico 'Dr. Eduardo Liceaga', 06720 Mexico City, Mexico
| | - Rafael Villalobos-Molina
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Mexico.,Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - José M Fragoso
- Departamento de Biología Molecular, Instituto Nacional de Cardiología 'Ignacio Chávez', Mexico City, Mexico
| | - Sonia León-Cabrera
- Carrera de Médico Cirujano, Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Avenida de los Barrios 1, Los Reyes Iztacala 54090, Mexico
| | - Pedro Ostoa-Saloma
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico
| | - Ruy Pérez-Tamayo
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Galileo Escobedo
- Laboratory for Proteomics & Metabolomics, Research Division, General Hospital of Mexico 'Dr. Eduardo Liceaga', 06720 Mexico City, Mexico
| |
Collapse
|
24
|
Ho KV, Schreiber KL, Vu DC, Rottinghaus SM, Jackson DE, Brown CR, Lei Z, Sumner LW, Coggeshall MV, Lin CH. Black Walnut ( Juglans nigra) Extracts Inhibit Proinflammatory Cytokine Production From Lipopolysaccharide-Stimulated Human Promonocytic Cell Line U-937. Front Pharmacol 2019; 10:1059. [PMID: 31607915 PMCID: PMC6761373 DOI: 10.3389/fphar.2019.01059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 08/20/2019] [Indexed: 01/01/2023] Open
Abstract
Black walnut (Juglans nigra L.) is an excellent source of health-promoting compounds. Consumption of black walnuts has been linked to many health benefits (e.g., anti-inflammatory) stemming from its phytochemical composition and medicinal properties, but these effects have not been systematically studied or characterized. In this study, potential anti-inflammatory compounds found in kernel extracts of 10 black walnut cultivars were putatively identified using a metabolomic profiling analysis, revealing differences in potential anti-inflammatory capacities among examined cultivars. Five cultivars were examined for activities in the human promonocytic cell line U-937 by evaluating the effects of the extracts on the expression of six human inflammatory cytokines/chemokines using a bead-based, flow cytometric multiplex assay. The methanolic extracts of these cultivars were added at four concentrations (0.1, 0.3, 1, and 10 mg/ml) either before and after the addition of lipopolysaccharide (LPS) to human U-937 cells to examine their effect on cytokine production. Results from cytotoxicity and viability assays revealed that the kernel extracts had no toxic effect on the U-937 cells. Of the 13 cytokines [interleukin (IL)-1β, tumor necrosis factor alpha (TNF-α), monocyte chemoattractant protein (MCP)-1, IL-6, IL-8, IL-10, IL-12, IL-17, IL-18, IL-23, IL-33, interferon (IFN)-α, IFN-γ] measured, only six were detected under the culture conditions. The production of the six detected cytokines by phorbol 12-myristate 13-acetate (PMA)-differentiated, LPS-stimulated U-937 was significantly inhibited by the kernel extracts from two cultivars Surprise and Sparrow when the extracts were added before the addition of LPS. Other cultivars (Daniel, Mystry, and Sparks) showed weak or no significant effects on cytokine production. In contrast, no inhibitory effect was observed on the production of cytokines by PMA-differentiated, LPS-stimulated U-937 when the kernel extracts were added after the addition of LPS. The findings suggest that the extracts from certain black walnut cultivars, such as Sparrow and Surprise, are promising biological candidates for potentially decreasing the severity of inflammatory disease.
Collapse
Affiliation(s)
- Khanh-Van Ho
- Center for Agroforestry, School of Natural Resources, University of Missouri, Columbia, MO, United States.,Department of Food Technology, Can Tho University, Can Tho, Vietnam
| | - Kathy L Schreiber
- Cell and Immunobiology Core, University of Missouri, Columbia, MO, United States
| | - Danh C Vu
- Center for Agroforestry, School of Natural Resources, University of Missouri, Columbia, MO, United States
| | - Susan M Rottinghaus
- Cell and Immunobiology Core, University of Missouri, Columbia, MO, United States
| | - Daniel E Jackson
- Cell and Immunobiology Core, University of Missouri, Columbia, MO, United States
| | - Charles R Brown
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Zhentian Lei
- Metabolomics Center, University of Missouri, Columbia, MO, United States.,Department of Biochemistry, Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Lloyd W Sumner
- Metabolomics Center, University of Missouri, Columbia, MO, United States.,Department of Biochemistry, Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
| | - Mark V Coggeshall
- United States Northern Research Station, USDA-Forest Service, West Lafayette, IN, United States
| | - Chung-Ho Lin
- Center for Agroforestry, School of Natural Resources, University of Missouri, Columbia, MO, United States
| |
Collapse
|
25
|
Avdeeva AS, Cherkasova MV, Kusevich DA, Rybakova VV, Artyuhov AS, Dashinimaev EB, Chichasova NV, Nasonov EL. [The role of laboratory biomarkers in monitoring of rituximab biosimilar therapy (Acellbia, "BIOCAD") in patients with rheumatoid arthritis]. TERAPEVT ARKH 2019; 91:26-33. [PMID: 32598673 DOI: 10.26442/00403660.2019.05.000230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Indexed: 11/22/2022]
Abstract
AIM to evaluate the role of laboratory biomarkers in monitoring effectiveness of rituximab (RTM) biosimilar therapy in a total dose of 1200 mg. MATERIALS AND METHODS 20 patients (pts) with rheumatoid arthritis (RA) (18 woman, mean age 61.5(54-66.5) years, mean disease duration 39.5(20-84) months, mean DAS28 5.6(4.9-6.8)) received two intravenous RTM biosimilar infusions (600 mg №2) in combination with DMARDs and glucocorticoids. Laboratory biomarkers were assessed at baseline and weeks 12 and 24 after the first infusion of RTX. RESULTS RTM biosimilar induced decreases in DAS28, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP) at week 12 and 24, p.
Collapse
Affiliation(s)
- A S Avdeeva
- V.A. Nasonova Scientific and Research Institute of Rheumatology
| | - M V Cherkasova
- V.A. Nasonova Scientific and Research Institute of Rheumatology
| | - D A Kusevich
- I.M. Sechenov First Moscow State Medical University of Ministry of Health of the Russian Federation (Sechenov University)
| | - V V Rybakova
- I.M. Sechenov First Moscow State Medical University of Ministry of Health of the Russian Federation (Sechenov University)
| | - A S Artyuhov
- Department of Regenerative Medicine.,Institute of Developmental Biology, Russian Academy of Sciences, Laboratory of Cell Proliferation, Pirogov Russian National Research Medical University
| | - E B Dashinimaev
- Department of Regenerative Medicine.,Institute of Developmental Biology, Russian Academy of Sciences, Laboratory of Cell Proliferation, Pirogov Russian National Research Medical University
| | - N V Chichasova
- V.A. Nasonova Scientific and Research Institute of Rheumatology.,I.M. Sechenov First Moscow State Medical University of Ministry of Health of the Russian Federation (Sechenov University)
| | - E L Nasonov
- V.A. Nasonova Scientific and Research Institute of Rheumatology.,I.M. Sechenov First Moscow State Medical University of Ministry of Health of the Russian Federation (Sechenov University)
| |
Collapse
|
26
|
Liao LC, Hu B, Zhang SP. Macrophages participate in the immunosuppression of condyloma acuminatum through the PD-1/PD-L1 signaling pathway. J Chin Med Assoc 2019; 82:413-418. [PMID: 30893264 DOI: 10.1097/jcma.0000000000000090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The aim of this study was to investigate whether macrophages can participate in the immunosuppression of condyloma acuminatum (CA) by expressing PD-1/PD-L1. METHODS The infiltration of macrophages and expression of programmed death-1 (PD-1) and PD-1 ligand 1 (PD-L1) on wart lesions and in normal skin tissues were detected by immunohistochemistry assay. The amounts of M1- and M2-type macrophages derived from THP-1 cells were measured by flow cytometry. The expression of cytokines on macrophages was examined by using enzyme-linked immunosorbent assay (ELISA). The protein expression of PD-1 and PD-L1 on macrophages was detected by western blotting. RESULTS The macrophages were significantly increased, while PD-1 and PD-L1 were highly expressed on wart lesions compared to normal controls. More M2-like macrophages than M1-type macrophages were present on wart lesions. The M2-like macrophages in the CA groups showed high expression of interleukin-10 (IL-10), transforming growth factor (TGF-β), PD-1, and PD-L1 compared to normal controls. CONCLUSION Macrophages participate in the immunosuppression of CA by expressing PD-1/PD-L1.
Collapse
|
27
|
Minshawi F, White MRH, Muller W, Humphreys N, Jackson D, Campbell BJ, Adamson A, Papoutsopoulou S. Human TNF-Luc reporter mouse: A new model to quantify inflammatory responses. Sci Rep 2019; 9:193. [PMID: 30655563 PMCID: PMC6336827 DOI: 10.1038/s41598-018-36969-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/19/2018] [Indexed: 12/26/2022] Open
Abstract
Tumour necrosis factor (TNF) is a key cytokine during inflammatory responses and its dysregulation is detrimental in many inflammatory diseases, such as rheumatoid arthritis and inflammatory bowel disease. Here, we used a bacterial artificial chromosome (BAC) construct that expresses luciferase under the control of the human TNF locus to generate a novel transgenic mouse, the hTNF.LucBAC strain. In vitro stimulation of hTNF.LucBAC cells of different origin revealed a cell specific response to stimuli demonstrating the integrated construct's ability as a proxy for inflammatory gene response. Lipopolysaccharide was the most potent luciferase inducer in macrophages, while TNF was a strong activator in intestinal organoids. Lipopolysaccharide-induced luciferase activity in macrophages was downregulated by inhibitors of NF-κB pathway, as well as by Interleukin-10, a known anti-inflammatory cytokine. Moreover, the transgene-dependent luciferase activity showed a positive correlation to the endogenous murine soluble TNF secreted to the culture medium. In conclusion, the hTNF.LucBAC strain is a valuable tool for studying and screening molecules that target TNF synthesis and will allow further functional studies of the regulatory elements of the TNF locus.
Collapse
Affiliation(s)
- Faisal Minshawi
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom.,Department of Laboratory Medicine, Faculty of Applied Medical Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mike R H White
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom
| | - Werner Muller
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom
| | - Neil Humphreys
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom
| | - Dean Jackson
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom
| | - Barry J Campbell
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GE, United Kingdom
| | - Antony Adamson
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom.
| | - Stamatia Papoutsopoulou
- Faculty of Biology, Medicine and Health, School of Biological Sciences, Manchester, M13 9PT, United Kingdom. .,Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3GE, United Kingdom.
| |
Collapse
|
28
|
Role of microRNA in severe asthma. Respir Investig 2018; 57:9-19. [PMID: 30455067 DOI: 10.1016/j.resinv.2018.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 12/23/2022]
Abstract
The various roles of microRNAs (miRNAs) in the epigenetic regulation of human disease are gaining importance as areas of research, and a better understanding of these roles may identify targets for development of novel therapies for severe asthma. MiRNAs, a class of small non-coding RNAs that serve as post-transcriptional gene repressors, are recognized as critical components in regulating tissue homeostasis. Alteration in miRNA expression disrupts homeostasis and is an underlying mechanism for development of chronic respiratory diseases, including asthma. Differential profiles of miRNA expression are involved in inflammation and remodeling pathogenicity via activating airway structural cells and immune cells and inducing cytokine releases. miRNA action leads to asthma progression from mild to severe stages. Here, current knowledge of the heterogeneous roles of miRNAs in severe asthma, including biological mechanisms underlying Th2 and macrophage polarization, type 2 innate lymphoid cell (ILC2) biology regulation, steroid-resistant asthma phenotype, airway smooth muscle (ASM) dysfunction, and impaired anti-viral innate immune, are reviewed.
Collapse
|
29
|
A novel fluorinated triazole derivative suppresses macrophage activation and alleviates experimental colitis via a Twist1-dependent pathway. Biochem Pharmacol 2018; 155:275-287. [DOI: 10.1016/j.bcp.2018.07.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/16/2018] [Indexed: 02/01/2023]
|
30
|
Physalin B Suppresses Inflammatory Response to Lipopolysaccharide in RAW264.7 Cells by Inhibiting NF-κB Signaling. J CHEM-NY 2018. [DOI: 10.1155/2018/7943140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Physalin B from Physalis angulata L. (Solanaceae) is a naturally occurring secosteroid with multiple biological activities. But its anti-inflammatory activity and mechanism remain unclear. Physalin B effects on RAW264.7 macrophages stimulated by lipopolysaccharide (LPS) were observed in this study. The expression and secretion of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) induced by LPS were significantly inhibited by physalin B. Meanwhile, the NF-κB nuclear translocation induced by LPS was inhibited by physalin B. The anti-inflammatory effects of physalin B could not be inhibited by mifepristone (RU486), the blocker of glucocorticoid receptor. In conclusion, physalin B can suppress inflammatory response to LPS in macrophages by inhibiting the production of inflammatory cytokines via NF-κB signaling.
Collapse
|
31
|
Page TH, Urbaniak AM, Espirito Santo AI, Danks L, Smallie T, Williams LM, Horwood NJ. Bruton's tyrosine kinase regulates TLR7/8-induced TNF transcription via nuclear factor-κB recruitment. Biochem Biophys Res Commun 2018; 499:260-266. [PMID: 29567473 PMCID: PMC5887515 DOI: 10.1016/j.bbrc.2018.03.140] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 03/19/2018] [Indexed: 02/05/2023]
Abstract
Tumour necrosis factor (TNF) is produced by primary human macrophages in response to stimulation by exogenous pathogen-associated molecular patterns (PAMPs) and endogenous damage-associated molecular patterns (DAMPs) via Toll-like receptor (TLR) signalling. However, uncontrolled TNF production can be deleterious and hence it is tightly controlled at multiple stages. We have previously shown that Bruton's tyrosine kinase (Btk) regulates TLR4-induced TNF production via p38 MAP Kinase by stabilising TNF messenger RNA. Using both gene over-expression and siRNA-mediated knockdown we have examined the role of Btk in TLR7/8 mediated TNF production. Our data shows that Btk acts in the TLR7/8 pathway and mediates Ser-536 phosphorylation of p65 RelA and subsequent nuclear entry in primary human macrophages. These data show an important role for Btk in TLR7/8 mediated TNF production and reveal distinct differences for Btk in TLR4 versus TLR7/8 signalling. Btk is required for TLR7/8 signalling in primary human macrophages. R848-induced TNF mRNA is more Btk dependent than LPS-induced TNF mRNA. Btk transcriptional control of TNF following R848 requires the promoter and 3′UTR. Btk knockdown reduces p65RelA translocation to the nucleus upon TLR7/8 stimulation.
Collapse
Affiliation(s)
- Theresa H Page
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, United Kingdom
| | - Anna M Urbaniak
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, United Kingdom
| | - Ana I Espirito Santo
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, United Kingdom
| | - Lynett Danks
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, United Kingdom
| | - Timothy Smallie
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, United Kingdom
| | - Lynn M Williams
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, United Kingdom
| | - Nicole J Horwood
- Kennedy Institute of Rheumatology, NDORMS, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, United Kingdom.
| |
Collapse
|
32
|
Abstract
Adipose tissue is a special tissue environment due to its high lipid content. Adipose tissue macrophages (ATMs) help maintain adipose tissue homeostasis in steady state by clearing dead adipocytes. However, adipose tissue changes drastically during obesity, resulting in a state of chronic low grade inflammation and a shift in the adipose immune landscape. In this review we will discuss how these changes influence the polarization of ATMs.
Collapse
Affiliation(s)
- Leen Catrysse
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.
| |
Collapse
|
33
|
Saradna A, Do DC, Kumar S, Fu QL, Gao P. Macrophage polarization and allergic asthma. Transl Res 2018; 191:1-14. [PMID: 29066321 PMCID: PMC5776696 DOI: 10.1016/j.trsl.2017.09.002] [Citation(s) in RCA: 280] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/13/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022]
Abstract
Allergic asthma is associated with airway inflammation and airway hyperresponsiveness. Macrophage polarization has been shown to have a profound impact on asthma pathogenesis. On exposure to local microenvironments, recruited macrophages can be polarized into either classically activated (or M1) or alternatively activated (or M2) phenotypes. Macrophage polarization has been heavily associated with development of asthma. The process of regulation of macrophage polarization involves an intricate interplay between various cytokines, chemokines, transcriptional factors, and immune-regulatory cells. Different signals from the microenvironment are controlled by different receptors on the macrophages to initiate various macrophage polarization pathways. Most importantly, there is an increased attention on the epigenetic changes (eg, microRNAs, DNA methylation, and histone modification) that impact macrophage functional responses and M1/M2 polarization through modulating cellular signaling and signature gene expression. Thus, modulation of macrophage phenotypes through molecular intervention by targeting some of those potential macrophage regulators may have therapeutic potential in the treatment of allergic asthma and other allergic diseases. In this review, we will discuss the origin of macrophages, characterization of macrophages, macrophage polarization in asthma, and the underlying mechanisms regarding allergen-induced macrophage polarization with emphasis on the regulation of epigenetics, which will provide new insights into the therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Arjun Saradna
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Department of Internal Medicine, Maimonides Medical Center, Brooklyn, NY
| | - Danh C Do
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Shruthi Kumar
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Bangalore Medical College and Research Institute, Bangalore, India
| | - Qing-Ling Fu
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peisong Gao
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
34
|
Jayesh K, Helen LR, Vysakh A, Binil E, Latha M. Ethyl acetate fraction of Terminalia bellirica (Gaertn.) Roxb. fruits inhibits proinflammatory mediators via down regulating nuclear factor-κB in LPS stimulated Raw 264.7 cells. Biomed Pharmacother 2017; 95:1654-1660. [DOI: 10.1016/j.biopha.2017.09.080] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/13/2017] [Accepted: 09/18/2017] [Indexed: 12/15/2022] Open
|
35
|
Tang T, Scambler TE, Smallie T, Cunliffe HE, Ross EA, Rosner DR, O'Neil JD, Clark AR. Macrophage responses to lipopolysaccharide are modulated by a feedback loop involving prostaglandin E 2, dual specificity phosphatase 1 and tristetraprolin. Sci Rep 2017; 7:4350. [PMID: 28659609 PMCID: PMC5489520 DOI: 10.1038/s41598-017-04100-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/09/2017] [Indexed: 01/02/2023] Open
Abstract
In many different cell types, pro-inflammatory agonists induce the expression of cyclooxygenase 2 (COX-2), an enzyme that catalyzes rate-limiting steps in the conversion of arachidonic acid to a variety of lipid signaling molecules, including prostaglandin E2 (PGE2). PGE2 has key roles in many early inflammatory events, such as the changes of vascular function that promote or facilitate leukocyte recruitment to sites of inflammation. Depending on context, it also exerts many important anti-inflammatory effects, for example increasing the expression of the anti-inflammatory cytokine interleukin 10 (IL-10), and decreasing that of the pro-inflammatory cytokine tumor necrosis factor (TNF). The tight control of both biosynthesis of, and cellular responses to, PGE2 are critical for the precise orchestration of the initiation and resolution of inflammatory responses. Here we describe evidence of a negative feedback loop, in which PGE2 augments the expression of dual specificity phosphatase 1, impairs the activity of mitogen-activated protein kinase p38, increases the activity of the mRNA-destabilizing factor tristetraprolin, and thereby inhibits the expression of COX-2. The same feedback mechanism contributes to PGE2-mediated suppression of TNF release. Engagement of the DUSP1-TTP regulatory axis by PGE2 is likely to contribute to the switch between initiation and resolution phases of inflammation.
Collapse
Affiliation(s)
- Tina Tang
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK
| | - Thomas E Scambler
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK
| | - Tim Smallie
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK
| | - Helen E Cunliffe
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK
| | - Ewan A Ross
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK
| | - Dalya R Rosner
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK
| | - John D O'Neil
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK
| | - Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2WB, UK.
| |
Collapse
|
36
|
Gain-of-Function Mutation of Tristetraprolin Impairs Negative Feedback Control of Macrophages In Vitro yet Has Overwhelmingly Anti-Inflammatory Consequences In Vivo. Mol Cell Biol 2017; 37:MCB.00536-16. [PMID: 28265004 PMCID: PMC5440651 DOI: 10.1128/mcb.00536-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 02/25/2017] [Indexed: 12/20/2022] Open
Abstract
The mRNA-destabilizing factor tristetraprolin (TTP) binds in a sequence-specific manner to the 3' untranslated regions of many proinflammatory mRNAs and recruits complexes of nucleases to promote rapid mRNA turnover. Mice lacking TTP develop a severe, spontaneous inflammatory syndrome characterized by the overexpression of tumor necrosis factor and other inflammatory mediators. However, TTP also employs the same mechanism to inhibit the expression of the potent anti-inflammatory cytokine interleukin 10 (IL-10). Perturbation of TTP function may therefore have mixed effects on inflammatory responses, either increasing or decreasing the expression of proinflammatory factors via direct or indirect mechanisms. We recently described a knock-in mouse strain in which the substitution of 2 amino acids of the endogenous TTP protein renders it constitutively active as an mRNA-destabilizing factor. Here we investigate the impact on the IL-10-mediated anti-inflammatory response. It is shown that the gain-of-function mutation of TTP impairs IL-10-mediated negative feedback control of macrophage function in vitro However, the in vivo effects of TTP mutation are uniformly anti-inflammatory despite the decreased expression of IL-10.
Collapse
|
37
|
Kumar Kingsley SM, Vishnu Bhat B. Role of MicroRNAs in the development and function of innate immune cells. Int Rev Immunol 2017; 36:154-175. [DOI: 10.1080/08830185.2017.1284212] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- S. Manoj Kumar Kingsley
- Department of Neonatology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| | - B. Vishnu Bhat
- Department of Neonatology, Jawaharlal Institute of Post Graduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
38
|
Worzfeld T, Pogge von Strandmann E, Huber M, Adhikary T, Wagner U, Reinartz S, Müller R. The Unique Molecular and Cellular Microenvironment of Ovarian Cancer. Front Oncol 2017; 7:24. [PMID: 28275576 PMCID: PMC5319992 DOI: 10.3389/fonc.2017.00024] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 02/07/2017] [Indexed: 12/13/2022] Open
Abstract
The reciprocal interplay of cancer cells and host cells is an indispensable prerequisite for tumor growth and progression. Cells of both the innate and adaptive immune system, in particular tumor-associated macrophages (TAMs) and T cells, as well as cancer-associated fibroblasts enter into a malicious liaison with tumor cells to create a tumor-promoting and immunosuppressive tumor microenvironment (TME). Ovarian cancer, the most lethal of all gynecological malignancies, is characterized by a unique TME that enables specific and efficient metastatic routes, impairs immune surveillance, and mediates therapy resistance. A characteristic feature of the ovarian cancer TME is the role of resident host cells, in particular activated mesothelial cells, which line the peritoneal cavity in huge numbers, as well as adipocytes of the omentum, the preferred site of metastatic lesions. Another crucial factor is the peritoneal fluid, which enables the transcoelomic spread of tumor cells to other pelvic and peritoneal organs, and occurs at more advanced stages as a malignancy-associated effusion. This ascites is rich in tumor-promoting soluble factors, extracellular vesicles and detached cancer cells as well as large numbers of T cells, TAMs, and other host cells, which cooperate with resident host cells to support tumor progression and immune evasion. In this review, we summarize and discuss our current knowledge of the cellular and molecular interactions that govern this interplay with a focus on signaling networks formed by cytokines, lipids, and extracellular vesicles; the pathophysiologial roles of TAMs and T cells; the mechanism of transcoelomic metastasis; and the cell type selective processing of signals from the TME.
Collapse
Affiliation(s)
- Thomas Worzfeld
- Institute of Pharmacology, Biochemical-Pharmacological Center (BPC), Philipps University, Marburg, Germany; Department of Pharmacology, Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Elke Pogge von Strandmann
- Experimental Tumor Research, Clinic for Hematology, Oncology and Immunology, Center for Tumor Biology and Immunology, Philipps University , Marburg , Germany
| | - Magdalena Huber
- Institute of Medical Microbiology and Hygiene, Biomedical Research Center, Philipps University , Marburg , Germany
| | - Till Adhikary
- Institute of Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University , Marburg , Germany
| | - Uwe Wagner
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, University Hospital of Giessen and Marburg (UKGM) , Marburg , Germany
| | - Silke Reinartz
- Clinic for Gynecology, Gynecological Oncology and Gynecological Endocrinology, Center for Tumor Biology and Immunology (ZTI), Philipps University , Marburg , Germany
| | - Rolf Müller
- Institute of Molecular Biology and Tumor Research, Center for Tumor Biology and Immunology, Philipps University , Marburg , Germany
| |
Collapse
|
39
|
Yang YJ, Yi L, Wang Q, Xie BB, Dong Y, Sha CW. Anti-inflammatory effects of physalin E from Physalis angulata on lipopolysaccharide-stimulated RAW 264.7 cells through inhibition of NF-κB pathway. Immunopharmacol Immunotoxicol 2017; 39:74-79. [DOI: 10.1080/08923973.2017.1282514] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Yan-Jun Yang
- Guangdong Food and Drug Vocational College, Guangdong Institute of Chinese Materia Medica, Guangzhou, China
| | - Lang Yi
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qing Wang
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bing-Bing Xie
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yan Dong
- Department of Immunology, Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cong-Wei Sha
- Guangdong Provincial Institute of Biological Products and Materia Medica, Guangzhou, China
| |
Collapse
|
40
|
Acetyl-L-Carnitine via Upegulating Dopamine D1 Receptor and Attenuating Microglial Activation Prevents Neuronal Loss and Improves Memory Functions in Parkinsonian Rats. Mol Neurobiol 2016; 55:583-602. [PMID: 27975173 DOI: 10.1007/s12035-016-0293-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 11/08/2016] [Indexed: 01/22/2023]
Abstract
Parkinson's disease is accompanied by nonmotor symptoms including cognitive impairment, which precede the onset of motor symptoms in patients and are regulated by dopamine (DA) receptors and the mesocorticolimbic pathway. The relative contribution of DA receptors and astrocytic glutamate transporter (GLT-1) in cognitive functions is largely unexplored. Similarly, whether microglia-derived increased immune response affects cognitive functions and neuronal survival is not yet understood. We have investigated the effect of acetyl-L-carnitine (ALCAR) on cognitive functions and its possible underlying mechanism of action in 6-hydroxydopamine (6-OHDA)-induced hemiparkinsonian rats. ALCAR treatment in 6-OHDA-lesioned rats improved memory functions as confirmed by decreased latency time and path length in the Morris water maze test. ALCAR further enhanced D1 receptor levels without altering D2 receptor levels in the hippocampus and prefrontal cortex (PFC) regions, suggesting that the D1 receptor is preferentially involved in the regulation of cognitive functions. ALCAR attenuated microglial activation and release of inflammatory mediators through balancing proinflammatory and anti-inflammatory cytokines, which subsequently enhanced the survival of mature neurons in the CA1, CA3, and PFC regions and improved cognitive functions in hemiparkinsonian rats. ALCAR treatment also improved glutathione (GSH) content, while decreasing oxidative stress indices, inducible nitrogen oxide synthase (iNOS) levels, and astrogliosis resulting in the upregulation of GLT-1 levels. Additionally, ALCAR prevented the loss of dopaminergic (DAergic) neurons in ventral tagmental area (VTA)/substantia nigra pars compacta (SNpc) regions of 6-OHDA-lesioned rats, thus maintaining the integrity of the nigrostriatal pathway. Together, these results demonstrate that ALCAR treatment in hemiparkinsonian rats ameliorates neurodegeneration and cognitive deficits, hence suggesting its therapeutic potential in neurodegenerative diseases.
Collapse
|
41
|
Shikonin inhibits TNF-α production through suppressing PKC-NF-κB-dependent decrease of IL-10 in rheumatoid arthritis-like cell model. J Nat Med 2016; 71:349-356. [DOI: 10.1007/s11418-016-1064-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/30/2016] [Indexed: 10/20/2022]
|
42
|
Hoodless LJ, Lucas CD, Duffin R, Denvir MA, Haslett C, Tucker CS, Rossi AG. Genetic and pharmacological inhibition of CDK9 drives neutrophil apoptosis to resolve inflammation in zebrafish in vivo. Sci Rep 2016; 5:36980. [PMID: 27833165 PMCID: PMC5105078 DOI: 10.1038/srep36980] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/24/2016] [Indexed: 01/12/2023] Open
Abstract
Neutrophilic inflammation is tightly regulated and subsequently resolves to limit tissue damage and promote repair. When the timely resolution of inflammation is dysregulated, tissue damage and disease results. One key control mechanism is neutrophil apoptosis, followed by apoptotic cell clearance by phagocytes such as macrophages. Cyclin-dependent kinase (CDK) inhibitor drugs induce neutrophil apoptosis in vitro and promote resolution of inflammation in rodent models. Here we present the first in vivo evidence, using pharmacological and genetic approaches, that CDK9 is involved in the resolution of neutrophil-dependent inflammation. Using live cell imaging in zebrafish with labelled neutrophils and macrophages, we show that pharmacological inhibition, morpholino-mediated knockdown and CRISPR/cas9-mediated knockout of CDK9 enhances inflammation resolution by reducing neutrophil numbers via induction of apoptosis after tailfin injury. Importantly, knockdown of the negative regulator La-related protein 7 (LaRP7) increased neutrophilic inflammation. Our data show that CDK9 is a possible target for controlling resolution of inflammation.
Collapse
Affiliation(s)
- Laura J. Hoodless
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Christopher D. Lucas
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Rodger Duffin
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Martin A. Denvir
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Christopher Haslett
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Carl S. Tucker
- BHF Centre for Cardiovascular Science, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| | - Adriano G. Rossi
- MRC Centre for Inflammation Research, The Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| |
Collapse
|
43
|
Coordinated Regulation of Signaling Pathways during Macrophage Activation. Microbiol Spectr 2016; 4. [PMID: 27780013 DOI: 10.1128/microbiolspec.mchd-0025-2015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The functional and phenotypic diversity of macrophages has long been appreciated, and it is now clear that it reflects a complex interplay between hard-wired differentiation pathways and instructive signals in specific tissues (Lawrence T, Natoli G. 2011, Nat Rev Immunol11:750-761). Recent studies have begun to unravel the molecular basis for the integration of these intrinsic developmental pathways with extracellular signals from the tissue microenvironment that confer the distinct phenotypes of tissue-resident macrophages (Lavin Y et al. 2014. Cell159:1312-1326; Gosselin D et al. 2014. Cell159:1327-1340). Macrophage phenotype and function is particularly dynamic during inflammation or infection, as blood monocytes are recruited into tissues and differentiate into macrophages, and depending on the nature of the inflammatory stimulus, they may acquire distinct functional phenotypes (Xue J et al. 2014. Immunity40:274-288; Murray PJ et al. 2014. Immunity41:14-20). Furthermore, these functional activation states can be rapidly modified in response to a changing microenvironment. Here we will discuss several key signaling pathways that drive macrophage activation during the inflammatory response and discuss how these pathways are integrated to "fine-tune" macrophage phenotype and function.
Collapse
|
44
|
Posttranscriptional control of NLRP3 inflammasome activation in colonic macrophages. Mucosal Immunol 2016; 9:850-8. [PMID: 26627461 PMCID: PMC4889550 DOI: 10.1038/mi.2015.109] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/29/2015] [Indexed: 02/04/2023]
Abstract
Colonic macrophages (cMPs) are important for intestinal homeostasis as they kill microbes and yet produce regulatory cytokines. Activity of the NLRP3 (nucleotide-binding leucine-rich repeat-containing pyrin receptor 3) inflammasome, a major sensor of stress and microorganisms that results in pro-inflammatory cytokine production and cell death, must be tightly controlled in the intestine. We demonstrate that resident cMPs are hyporesponsive to NLRP3 inflammasome activation owing to a remarkable level of posttranscriptional control of NLRP3 and pro-interleukin-1β (proIL-1β) protein expression, which was also seen for tumor necrosis factor-α and IL-6, but lost during experimental colitis. Resident cMPs rapidly degraded NLRP3 and proIL-1β proteins by the ubiquitin/proteasome system. Finally, blocking IL-10R-signaling in vivo enhanced NLRP3 and proIL-1β protein but not mRNA levels in resident cMPs, implicating a role for IL-10 in environmental conditioning of cMPs. These data are the first to show dramatic posttranscriptional control of inflammatory cytokine production by a relevant tissue-derived macrophage population and proteasomal degradation of proIL-1β and NLRP3 as a mechanism to control inflammasome activation, findings which have broad implications for our understanding of intestinal and systemic inflammatory diseases.
Collapse
|
45
|
Warming to 39°C but Not to 37°C Ameliorates the Effects on the Monocyte Response by Hypothermia. Ann Surg 2016; 263:601-7. [DOI: 10.1097/sla.0000000000001175] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
46
|
Chan JK, Glass GE, Ersek A, Freidin A, Williams GA, Gowers K, Espirito Santo AI, Jeffery R, Otto WR, Poulsom R, Feldmann M, Rankin SM, Horwood NJ, Nanchahal J. Low-dose TNF augments fracture healing in normal and osteoporotic bone by up-regulating the innate immune response. EMBO Mol Med 2016; 7:547-61. [PMID: 25770819 PMCID: PMC4492816 DOI: 10.15252/emmm.201404487] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mechanism by which trauma initiates healing remains unclear. Precise understanding of these events may define interventions for accelerating healing that could be translated to the clinical arena. We previously reported that addition of low-dose recombinant human TNF (rhTNF) at the fracture site augmented fracture repair in a murine tibial fracture model. Here, we show that local rhTNF treatment is only effective when administered within 24 h of injury, when neutrophils are the major inflammatory cell infiltrate. Systemic administration of anti-TNF impaired fracture healing. Addition of rhTNF enhanced neutrophil recruitment and promoted recruitment of monocytes through CCL2 production. Conversely, depletion of neutrophils or inhibition of the chemokine receptor CCR2 resulted in significantly impaired fracture healing. Fragility, or osteoporotic, fractures represent a major medical problem as they are associated with permanent disability and premature death. Using a murine model of fragility fractures, we found that local rhTNF treatment improved fracture healing during the early phase of repair. If translated clinically, this promotion of fracture healing would reduce the morbidity and mortality associated with delayed patient mobilization.
Collapse
Affiliation(s)
- James K Chan
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Graeme E Glass
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Adel Ersek
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Freidin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Garry A Williams
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Kate Gowers
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Rosemary Jeffery
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - William R Otto
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Richard Poulsom
- Histopathology Laboratory and In Situ Hybridisation Service, Cancer Research UK - London Research Institute, London, UK
| | - Marc Feldmann
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Sara M Rankin
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Nicole J Horwood
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | |
Collapse
|
47
|
Hoodless LJ, Robb CT, Felton JM, Tucker CS, Rossi AG. Models for the Study of the Cross Talk Between Inflammation and Cell Cycle. Methods Mol Biol 2016; 1336:179-209. [PMID: 26231717 DOI: 10.1007/978-1-4939-2926-9_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Cyclin-dependent kinases (CDKs) have been traditionally associated with the cell cycle. However, it is now known that CDK7 and CDK9 regulate transcriptional activity via phosphorylation of RNA polymerase II and subsequent synthesis of, for example, inflammatory mediators and factors that influence the apoptotic process; including apoptosis of granulocytes such as neutrophils and eosinophils. Successful resolution of inflammation and restoration of normal tissue homeostasis requires apoptosis of these inflammatory cells and subsequent clearance of apoptotic bodies by phagocytes such as macrophages. It is believed that CDK7 and CDK9 influence resolution of inflammation since they are involved in the transcription of anti-apoptotic proteins such as Mcl-1 which is especially important in granulocyte survival.This chapter describes various in vitro and in vivo models used to investigate CDKs and their inhibitors in granulocytes and particularly the role of CDKs in the apoptosis pathway. This can be performed in vitro by isolation and use of primary granulocytes and in vivo using animal models of inflammatory disease in rodents and zebrafish. Some of the methods described here to assess the role of CDKs in inflammation and apoptosis include flow cytometry and western blotting, together with imaging and quantification of apoptosis in fixed tissue, as well as in vivo models of inflammation.
Collapse
Affiliation(s)
- Laura J Hoodless
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | | | | | | | | |
Collapse
|
48
|
|
49
|
IL-10 disrupts the Brd4-docking sites to inhibit LPS-induced CXCL8 and TNF-α expression in monocytes: Implications for chronic obstructive pulmonary disease. J Allergy Clin Immunol 2015; 136:781-791.e9. [DOI: 10.1016/j.jaci.2015.04.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 03/30/2015] [Accepted: 04/13/2015] [Indexed: 11/18/2022]
|
50
|
To WS, Aungier SR, Cartwright AJ, Ito K, Midwood KS. Potent anti-inflammatory effects of the narrow spectrum kinase inhibitor RV1088 on rheumatoid arthritis synovial membrane cells. Br J Pharmacol 2015; 172:3805-16. [PMID: 25891413 PMCID: PMC4523337 DOI: 10.1111/bph.13170] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 03/29/2015] [Accepted: 04/08/2015] [Indexed: 12/29/2022] Open
Abstract
Background and Purpose To investigate whether a narrow spectrum kinase inhibitor RV1088, which simultaneously targets specific MAPKs, Src and spleen tyrosine kinase (Syk), is more effective at inhibiting inflammatory signalling in rheumatoid arthritis (RA) than single kinase inhibitors (SKIs). Experimental Approach elisas were used to determine the efficacy of RV1088, clinically relevant SKIs and the pharmaceutical Humira on pro-inflammatory cytokine production by activated RA synovial fibroblasts, primary human monocytes and macrophages, as well as spontaneous cytokine synthesis by synovial membrane cells from RA patients. In human macrophages, RNAi knockdown of individual kinases was used to reveal the effect of inhibition of kinase expression on cytokine synthesis. Key Results RV1088 reduced TNF-α, IL-6 and IL-8 production in all individual activated cell types with low, nM, IC50s. SKIs, and combinations of SKIs, were significantly less effective than RV1088. RNAi of specific kinases in macrophages also caused only modest inhibition of pro-inflammatory cytokine production. RV1088 was also significantly more effective at inhibiting IL-6 and IL-8 production by monocytes and RA synovial fibroblasts compared with Humira. Finally, RV1088 was the only inhibitor that was effective in reducing TNF-α, IL-6 and IL-8 synthesis in RA synovial membrane cells with low nM IC50s. Conclusions and Implications This study demonstrates potent anti-inflammatory effect of RV1088, highlighting that distinct signalling pathways drive TNF-α, IL-6 and IL-8 production in the different cell types found in RA joints. As such, targeting numerous signalling pathways simultaneously using RV1088 could offer a more powerful method of reducing inflammation in RA than targeting individual kinases.
Collapse
Affiliation(s)
- Wing S To
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Susan R Aungier
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Alison J Cartwright
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Kazuhiro Ito
- Airway Disease, National Heart and Lung Institute, Imperial College, London, UK
| | - Kim S Midwood
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| |
Collapse
|