1
|
Sørhus E, Lie KK, Meier S, Furmanek T, Norberg B, Perrichon P. Transcriptomics uncover inhibition of repair and wound healing pathways in Atlantic halibut (Hippoglossus hippoglossus) after crude oil exposure. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118151. [PMID: 40222109 DOI: 10.1016/j.ecoenv.2025.118151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
Accidental oil spills significantly threaten marine ecosystems and fisheries, impacting biodiversity and ecological health. This study examines the downstream transcriptomic responses of Atlantic halibut larvae exposed to crude oil during organogenesis. Findings show concentration-dependent transcriptional abnormalities. Pathway analysis at 10 days post-hatch (dph), 11 days after cessation of oil exposure, indicates downregulation of inflammatory and reparative pathways. By 18 dph, tissue-specific analyses reveal activation of these pathways, especially in head tissues, alongside upregulation of neuronal signaling pathways. This highlights the complex relationship between oil exposure and transcriptional responses, emphasizing recovery mechanisms represented by regulation of inflammatory, repair and wound healing pathways following oil exposure. The activation of repair pathways in surviving larvae suggests compensatory processes to address oil-induced damage. These novel insights enhance understanding of the molecular mechanisms of oil toxicity and the lasting effects on marine organisms.
Collapse
Affiliation(s)
- Elin Sørhus
- Institute of Marine Research, Nordnesgaten 50, Bergen, Norway.
| | - Kai K Lie
- Institute of Marine Research, Nordnesgaten 50, Bergen, Norway
| | - Sonnich Meier
- Institute of Marine Research, Nordnesgaten 50, Bergen, Norway
| | - Tomasz Furmanek
- Institute of Marine Research, Nordnesgaten 50, Bergen, Norway
| | - Birgitta Norberg
- Institute of Marine Research, Austevoll Research station, Sauganeset 16, Storebø, Norway
| | - Prescilla Perrichon
- Institute of Marine Research, Austevoll Research station, Sauganeset 16, Storebø, Norway
| |
Collapse
|
2
|
Saveh-Shemshaki N, Barajaa MA, Otsuka T, Mirdamadi ES, Nair LS, Laurencin CT. Electroconductivity, a regenerative engineering approach to reverse rotator cuff muscle degeneration. Regen Biomater 2023; 10:rbad099. [PMID: 38020235 PMCID: PMC10676522 DOI: 10.1093/rb/rbad099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/25/2023] [Accepted: 10/28/2023] [Indexed: 12/01/2023] Open
Abstract
Muscle degeneration is one the main factors that lead to the high rate of retear after a successful repair of rotator cuff (RC) tears. The current surgical practices have failed to treat patients with chronic massive rotator cuff tears (RCTs). Therefore, regenerative engineering approaches are being studied to address the challenges. Recent studies showed the promising outcomes of electroactive materials (EAMs) on the regeneration of electrically excitable tissues such as skeletal muscle. Here, we review the most important biological mechanism of RC muscle degeneration. Further, the review covers the recent studies on EAMs for muscle regeneration including RC muscle. Finally, we will discuss the future direction toward the application of EAMs for the augmentation of RCTs.
Collapse
Affiliation(s)
- Nikoo Saveh-Shemshaki
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Mohammed A Barajaa
- Department of Biomedical Engineering, Imam Abdulrahman Bin Faisal University, Dammam 31451, Saudi Arabia
| | - Takayoshi Otsuka
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
| | - Elnaz S Mirdamadi
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Lakshmi S Nair
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Cato T Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Farmington, CT 06030, USA
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Orthopedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
- Department of Chemical & Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
3
|
Orthobiologic Treatment of Ligament Injuries. Phys Med Rehabil Clin N Am 2023; 34:135-163. [DOI: 10.1016/j.pmr.2022.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
4
|
Jia D, Cai J, Yao F, Zhu P, Xu X, Qi Y, Wang H. Effect of Bacillus Subtilis on Immune Function of Hd11 Chicken Macrophages. BRAZILIAN JOURNAL OF POULTRY SCIENCE 2023. [DOI: 10.1590/1806-9061-2022-1641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Affiliation(s)
- D Jia
- Jiangsu Lihua Animal Husbandry Co., Ltd, P.R.China
| | - J Cai
- Yangzhou University, P.R.China
| | - F Yao
- Yangzhou University, P.R.China
| | - P Zhu
- Jiangsu Lihua Animal Husbandry Co., Ltd, P.R.China; Yangzhou University, P.R.China
| | - X Xu
- Jiangsu Lihua Animal Husbandry Co., Ltd, P.R.China
| | - Y Qi
- Jiangsu Lihua Animal Husbandry Co., Ltd, P.R.China
| | - H Wang
- Yangzhou University, P.R.China
| |
Collapse
|
5
|
Papathanasiou E, Scott AR, Trotman CA, Beale C, Price LL, Huggins GS, Zhang Y, Georgakoudi I, Van Dyke TE. Specialized Pro-Resolving Mediators Reduce Scarring After Cleft Lip Repair. Front Immunol 2022; 13:871200. [PMID: 35572588 PMCID: PMC9094441 DOI: 10.3389/fimmu.2022.871200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Residual scarring after cleft lip repair surgery remains a challenge for both surgeons and patients and novel therapeutics are critically needed. The objective of this preclinical experimental study was to evaluate the impact of the methyl-ester of pro-resolving lipid mediator lipoxin A4 (LXA4-ME) on scarring in a novel rabbit model of cleft lip repair. Methods A defect of the lip was surgically created and repaired in eight six-week old New Zealand white rabbits to simulate human cleft lip scars. Rabbits were randomly assigned to topical application of PBS (control) or 1 ug of LXA4-ME (treatment). 42 days post surgery all animals were euthanized. Photographs of the cleft lip area defect and histologic specimens were evaluated. Multiple scar assessment scales were used to compare scarring. Results Animals treated with LXA4-ME exhibited lower Visual Scar Assessment scores compared to animals treated with PBS. Treatment with LXA4-ME resulted in a significant reduction of inflammatory cell infiltrate and density of collagen fibers. Control animals showed reduced 2D directional variance (orientation) of collagen fibers compared to animals treated with LXA4-ME demonstrating thicker and more parallel collagen fibers, consistent with scar tissue. Conclusions These data suggest that LXA4-ME limits scarring after cleft lip repair and improves wound healing outcomes in rabbits favoring the resolution of inflammation. Further studies are needed to explore the mechanisms that underlie the positive therapeutic impact of LXA4-ME on scarring to set the stage for future human clinical trials of LXA4-ME for scar prevention or treatment after cleft lip repair.
Collapse
Affiliation(s)
- Evangelos Papathanasiou
- Department of Periodontology, Tufts University School of Dental Medicine, Boston, MA, United States
- Center for Clinical and Translational Research, Forsyth Institute, Cambridge, MA, United States
| | - Andrew R. Scott
- Department of Otolaryngology – Head & Neck Surgery, Tufts University School of Medicine, Boston, MA, United States
| | - Carroll Ann Trotman
- College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Corinna Beale
- Tufts Comparative Medicine Services, Tufts University, Boston, MA, United States
| | - Lori Lyn Price
- Tufts Clinical and Translational Science Institute, Tufts University, Boston, MA, United States
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, United States
| | - Gordon S. Huggins
- Molecular Cardiology Research Institute and Cardiology Division, Tufts Medical Center and Tufts University School of Medicine, Boston, MA, United States
| | - Yang Zhang
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA, United States
| | - Irene Georgakoudi
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, MA, United States
| | - Thomas E. Van Dyke
- Center for Clinical and Translational Research, Forsyth Institute, Cambridge, MA, United States
- Department of Oral Medicine, Infection and Immunity, Faculty of Medicine, Harvard University, Boston, MA, United States
| |
Collapse
|
6
|
Russo V, El Khatib M, Prencipe G, Citeroni MR, Faydaver M, Mauro A, Berardinelli P, Cerveró-Varona A, Haidar-Montes AA, Turriani M, Di Giacinto O, Raspa M, Scavizzi F, Bonaventura F, Stöckl J, Barboni B. Tendon Immune Regeneration: Insights on the Synergetic Role of Stem and Immune Cells during Tendon Regeneration. Cells 2022; 11:434. [PMID: 35159244 PMCID: PMC8834336 DOI: 10.3390/cells11030434] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/11/2022] Open
Abstract
Tendon disorders represent a very common pathology in today's population, and tendinopathies that account 30% of tendon-related injuries, affect yearly millions of people which in turn cause huge socioeconomic and health repercussions worldwide. Inflammation plays a prominent role in the development of tendon pathologies, and advances in understanding the underlying mechanisms during the inflammatory state have provided additional insights into its potential role in tendon disorders. Different cell compartments, in combination with secreted immune modulators, have shown to control and modulate the inflammatory response during tendinopathies. Stromal compartment represented by tenocytes has shown to display an important role in orchestrating the inflammatory response during tendon injuries due to the interplay they exhibit with the immune-sensing and infiltrating compartments, which belong to resident and recruited immune cells. The use of stem cells or their derived secretomes within the regenerative medicine field might represent synergic new therapeutical approaches that can be used to tune the reaction of immune cells within the damaged tissues. To this end, promising opportunities are headed to the stimulation of macrophages polarization towards anti-inflammatory phenotype together with the recruitment of stem cells, that possess immunomodulatory properties, able to infiltrate within the damaged tissues and improve tendinopathies resolution. Indeed, the comprehension of the interactions between tenocytes or stem cells with the immune cells might considerably modulate the immune reaction solving hence the inflammatory response and preventing fibrotic tissue formation. The purpose of this review is to compare the roles of distinct cell compartments during tendon homeostasis and injury. Furthermore, the role of immune cells in this field, as well as their interactions with stem cells and tenocytes during tendon regeneration, will be discussed to gain insights into new ways for dealing with tendinopathies.
Collapse
Affiliation(s)
- Valentina Russo
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Mohammad El Khatib
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maria Rita Citeroni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Melisa Faydaver
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Annunziata Mauro
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Arlette A. Haidar-Montes
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Maura Turriani
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Oriana Di Giacinto
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| | - Marcello Raspa
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Ferdinando Scavizzi
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Fabrizio Bonaventura
- National Research Council (CNR), Campus International Development (EMMA-INFRAFRONTIER-IMPC), Institute of Biochemistry and Cellular Biology (IBBC), 00015 Monterotondo Scalo, Italy; (M.R.); (F.S.); (F.B.)
| | - Johannes Stöckl
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Faculty of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy; (V.R.); (M.E.K.); (M.R.C.); (M.F.); (A.M.); (P.B.); (A.C.-V.); (A.A.H.-M.); (M.T.); (O.D.G.); (B.B.)
| |
Collapse
|
7
|
Giese AA, Babendreyer A, Krappen P, Gross A, Strnad P, Düsterhöft S, Ludwig A. Inflammatory activation of surface molecule shedding by upregulation of the pseudoprotease iRhom2 in colon epithelial cells. Sci Rep 2021; 11:24230. [PMID: 34930929 PMCID: PMC8688420 DOI: 10.1038/s41598-021-03522-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/26/2021] [Indexed: 01/09/2023] Open
Abstract
The metalloproteinase ADAM17 contributes to inflammatory and proliferative responses by shedding of cell-surface molecules. By this ADAM17 is implicated in inflammation, regeneration, and permeability regulation of epithelial cells in the colon. ADAM17 maturation and surface expression requires the adapter proteins iRhom1 or iRhom2. Here we report that expression of iRhom2 but not iRhom1 is upregulated in intestinal tissue of mice with acute colitis. Our analysis of public databases indicates elevated iRhom2 expression in mucosal tissue and epithelial cells from patients with inflammatory bowel disease (IBD). Consistently, expression of iRhom2 but not iRhom1 is upregulated in colon or intestinal epithelial cell lines after co-stimulation with tumor necrosis factor (TNF) and interferon gamma (IFNgamma). This upregulation can be reduced by inhibition of Janus kinases or transcription factors NF-kappaB or AP-1. Upregulation of iRhom2 can be mimicked by iRhom2 overexpression and is associated with enhanced maturation and surface expression of ADAM17 which then results in increased cleavage of transforming growth factor (TGF) alpha and junctional adhesion molecule (JAM)-A. Finally, the induction of these responses is suppressed by inhibition of iRhom2 transcription. Thus, inflammatory induction of iRhom2 may contribute to upregulated ADAM17-dependent mediator and adhesion molecule release in IBD. The development of iRhom2-dependent inhibitors may allow selective targeting of inflammatory ADAM17 activities.
Collapse
Affiliation(s)
- Anja Adelina Giese
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Aaron Babendreyer
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Peter Krappen
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Annika Gross
- Division of Gastroenterology and Hepatology, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Pavel Strnad
- Division of Gastroenterology and Hepatology, Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Andreas Ludwig
- Institute of Molecular Pharmacology, Medical Faculty, RWTH Aachen University, Pauwelsstr. 30, 52074, Aachen, Germany.
| |
Collapse
|
8
|
Kırıcı P, Kaplan S, Aydin Turk B, Annac E. Histopathological Examination of the Mucosal Effects of Obstetric Gel on Vaginal Wound Healing in an Incision-Inflicted Rat Model. Cureus 2021; 13:e18254. [PMID: 34712531 PMCID: PMC8542395 DOI: 10.7759/cureus.18254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2021] [Indexed: 11/05/2022] Open
Abstract
Background and objective The present study intended to investigate the histopathological efficacy of obstetric gels on the healing of vaginal lacerations in rats. To the best of our knowledge, this is the first such study. Materials and methods Twenty-one female Wistar albino rats were divided into three groups, comprising seven animals per group. The first group (group 1) was the control group, the second (group 2) was the polyvinyl iodine (PI) group, and the third group (group 3) was the obstetric gel (OG) group. In all three groups, a vaginal incision was made with a No. 10 scalpel, and the incision site was sutured with a 3-0 Vicryl suture. In the control group, the incision site was left for routine healing. The incision site was washed with PI in the PI group and with OG in the OG group. After 15 days, vaginal tissues were obtained from all three groups for histopathological examination. In addition, immunohistochemistry staining was performed using caspase 3 and fibrillin 1 antibodies. Results There was no significant difference between the groups in terms of congestion, vascular proliferation, and inflammation stages in the examinations performed on the vaginal wall. However, the amount of collagen and elastic fibers increased during the remodeling and fibrosis phase, and the fibrillin 1 score increased in immunohistochemistry staining (p < 0.001). Conclusion It has been shown in rat vaginal tissue that obstetric gels do not have negative effects on wound healing; however, they contribute to wound healing by positively affecting the fibrosis stage.
Collapse
Affiliation(s)
- Pınar Kırıcı
- Obstetrics and Gynaecology, Adiyaman University, Adıyaman, TUR
| | - Selçuk Kaplan
- Obstetrics and Gynaecology, Adiyaman University Faculty of Medicine, Adıyaman, TUR
| | - Bilge Aydin Turk
- Pathology, Adiyaman University Faculty of Medicine, Adıyaman, TUR
| | - Ebru Annac
- Histology and Embryology, Adiyaman University Faculty of Medicine, Adıyaman, TUR
| |
Collapse
|
9
|
Ross EA, Devitt A, Johnson JR. Macrophages: The Good, the Bad, and the Gluttony. Front Immunol 2021; 12:708186. [PMID: 34456917 PMCID: PMC8397413 DOI: 10.3389/fimmu.2021.708186] [Citation(s) in RCA: 246] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Macrophages are dynamic cells that play critical roles in the induction and resolution of sterile inflammation. In this review, we will compile and interpret recent findings on the plasticity of macrophages and how these cells contribute to the development of non-infectious inflammatory diseases, with a particular focus on allergic and autoimmune disorders. The critical roles of macrophages in the resolution of inflammation will then be examined, emphasizing the ability of macrophages to clear apoptotic immune cells. Rheumatoid arthritis (RA) is a chronic autoimmune-driven spectrum of diseases where persistent inflammation results in synovial hyperplasia and excessive immune cell accumulation, leading to remodeling and reduced function in affected joints. Macrophages are central to the pathophysiology of RA, driving episodic cycles of chronic inflammation and tissue destruction. RA patients have increased numbers of active M1 polarized pro-inflammatory macrophages and few or inactive M2 type cells. This imbalance in macrophage homeostasis is a main contributor to pro-inflammatory mediators in RA, resulting in continual activation of immune and stromal populations and accelerated tissue remodeling. Modulation of macrophage phenotype and function remains a key therapeutic goal for the treatment of this disease. Intriguingly, therapeutic intervention with glucocorticoids or other DMARDs promotes the re-polarization of M1 macrophages to an anti-inflammatory M2 phenotype; this reprogramming is dependent on metabolic changes to promote phenotypic switching. Allergic asthma is associated with Th2-polarised airway inflammation, structural remodeling of the large airways, and airway hyperresponsiveness. Macrophage polarization has a profound impact on asthma pathogenesis, as the response to allergen exposure is regulated by an intricate interplay between local immune factors including cytokines, chemokines and danger signals from neighboring cells. In the Th2-polarized environment characteristic of allergic asthma, high levels of IL-4 produced by locally infiltrating innate lymphoid cells and helper T cells promote the acquisition of an alternatively activated M2a phenotype in macrophages, with myriad effects on the local immune response and airway structure. Targeting regulators of macrophage plasticity is currently being pursued in the treatment of allergic asthma and other allergic diseases. Macrophages promote the re-balancing of pro-inflammatory responses towards pro-resolution responses and are thus central to the success of an inflammatory response. It has long been established that apoptosis supports monocyte and macrophage recruitment to sites of inflammation, facilitating subsequent corpse clearance. This drives resolution responses and mediates a phenotypic switch in the polarity of macrophages. However, the role of apoptotic cell-derived extracellular vesicles (ACdEV) in the recruitment and control of macrophage phenotype has received remarkably little attention. ACdEV are powerful mediators of intercellular communication, carrying a wealth of lipid and protein mediators that may modulate macrophage phenotype, including a cargo of active immune-modulating enzymes. The impact of such interactions may result in repair or disease in different contexts. In this review, we will discuss the origin, characterization, and activity of macrophages in sterile inflammatory diseases and the underlying mechanisms of macrophage polarization via ACdEV and apoptotic cell clearance, in order to provide new insights into therapeutic strategies that could exploit the capabilities of these agile and responsive cells.
Collapse
Affiliation(s)
- Ewan A Ross
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Andrew Devitt
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Jill R Johnson
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| |
Collapse
|
10
|
The pro-apoptotic ARTS protein induces neutrophil apoptosis, efferocytosis, and macrophage reprogramming to promote resolution of inflammation. Apoptosis 2021; 25:558-573. [PMID: 32564202 DOI: 10.1007/s10495-020-01615-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
ARTS (Sept4_i2) is a pro-apoptotic protein and a product of the Sept4 gene. ARTS acts upstream of mitochondria to initiate caspase activation. ARTS induces apoptosis by specifically binding XIAP and allowing de-repression of active caspases required for Mitochondrial Outer Membrane Permeabilzation (MOMP). Moreover, ARTS promotes apoptosis by inducing ubiquitin-mediated degradation of both major anti-apoptotic proteins XIAP and Bcl-2. In the resolution phase of inflammation, the infiltrating leukocytes, which execute the acute innate response, undergo apoptosis and are subsequently cleared by phagocytic macrophages (i.e. efferocytosis). In this course, macrophages undergo reprogramming from inflammatory, to anti-inflammatory, and eventually to resolving macrophages that leave the injury sites. Since engulfment of apoptotic leukocytes is a key signaling step in macrophage reprogramming and resolution of inflammation, we hypothesized that a failed apoptosis in leukocytes in vivo would result in an impaired resolution process. To test this hypothesis, we utilized the Sept4/ARTS-/- mice, which exhibit resistance to apoptosis in many cell types. During zymosan A-induced peritonitis, Sept4/ARTS-/- mice exhibited impaired resolution of inflammation, characterized by reduced neutrophil apoptosis, macrophage efferocytosis and expression of pro-resolving mediators. This was associated with increased pro-inflammatory cytokines and reduced anti-inflammatory cytokines, secreted by resolution-phase macrophages. Moreover, ARTS overexpression in leukocytes in vitro promoted an anti-inflammatory behavior. Overall, our results suggest that ARTS is a key master-regulator necessary for neutrophil apoptosis, macrophage efferocytosis and reprogramming to the pro-resolving phenotype during the resolution of inflammation.
Collapse
|
11
|
Li H, Yang Z, Fu L, Yuan Z, Gao C, Sui X, Liu S, Peng J, Dai Y, Guo Q. Advanced Polymer-Based Drug Delivery Strategies for Meniscal Regeneration. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:266-293. [PMID: 32988289 DOI: 10.1089/ten.teb.2020.0156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The meniscus plays a critical role in maintaining knee joint homeostasis. Injuries to the meniscus, especially considering the limited self-healing capacity of the avascular region, continue to be a challenge and are often treated by (partial) meniscectomy, which has been identified to cause osteoarthritis. Currently, meniscus tissue engineering focuses on providing extracellular matrix (ECM)-mimicking scaffolds to direct the inherent meniscal regeneration process, and it has been found that various stimuli are essential. Numerous bioactive factors present benefits in regulating cell fate, tissue development, and healing, but lack an optimal delivery system. More recently, bioengineers have developed various polymer-based drug delivery systems (PDDSs), which are beneficial in terms of the favorable properties of polymers as well as novel delivery strategies. Engineered PDDSs aim to provide not only an ECM-mimicking microenvironment but also the controlled release of bioactive factors with release profiles tailored according to the biological concerns and properties of the factors. In this review, both different polymers and bioactive factors involved in meniscal regeneration are discussed, as well as potential candidate systems, with examples of recent progress. This article aims to summarize drug delivery strategies in meniscal regeneration, with a focus on novel delivery strategies rather than on specific delivery carriers. The current challenges and future prospects for the structural and functional regeneration of the meniscus are also discussed. Impact statement Meniscal injury remains a clinical Gordian knot owing to the limited healing potential of the region, restricted surgical approaches, and risk of inducing osteoarthritis. Existing tissue engineering scaffolds that provide mechanical support and a favorable microenvironment also lack biological cues. Advanced polymer-based delivery strategies consisting of polymers incorporating bioactive factors have emerged as a promising direction. This article primarily reviews the types and applications of biopolymers and bioactive factors in meniscal regeneration. Importantly, various carrier systems and drug delivery strategies are discussed with the hope of inspiring further advancements in this field.
Collapse
Affiliation(s)
- Hao Li
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Zhen Yang
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Liwei Fu
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Zhiguo Yuan
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China.,Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Cangjian Gao
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Xiang Sui
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Shuyun Liu
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Jiang Peng
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| | - Yongjing Dai
- Department of Orthopedic, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, China.,Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital; Beijing Key Lab of Regenerative Medicine in Orthopedics; Key Laboratory of Musculoskeletal Trauma & War Injuries PLA; Beijing, China
| |
Collapse
|
12
|
Toko H, Morita H, Katakura M, Hashimoto M, Ko T, Bujo S, Adachi Y, Ueda K, Murakami H, Ishizuka M, Guo J, Zhao C, Fujiwara T, Hara H, Takeda N, Takimoto E, Shido O, Harada M, Komuro I. Omega-3 fatty acid prevents the development of heart failure by changing fatty acid composition in the heart. Sci Rep 2020; 10:15553. [PMID: 32968201 PMCID: PMC7512019 DOI: 10.1038/s41598-020-72686-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/04/2020] [Indexed: 12/22/2022] Open
Abstract
Some clinical trials showed that omega-3 fatty acid (FA) reduced cardiovascular events, but it remains unknown whether omega-3 FA supplementation changes the composition of FAs and their metabolites in the heart and how the changes, if any, exert beneficial effects on cardiac structure and function. To clarify these issues, we supplied omega-3 FA to mice exposed to pressure overload, and examined cardiac structure and function by echocardiography and a proportion of FAs and their metabolites by gas chromatography and liquid chromatography-tandem mass spectrometry, respectively. Pressure overload induced cardiac hypertrophy and dysfunction, and reduced concentration of all FAs’ components and increased free form arachidonic acid and its metabolites, precursors of pro-inflammatory mediators in the heart. Omega-3 FA supplementation increased both total and free form of eicosapentaenoic acid, a precursor of pro-resolution mediators and reduced free form arachidonic acid in the heart. Omega-3 FA supplementation suppressed expressions of pro-inflammatory cytokines and the infiltration of inflammatory cells into the heart and ameliorated cardiac dysfunction and fibrosis. These results suggest that omega-3 FA-induced changes of FAs composition in the heart have beneficial effects on cardiac function via regulating inflammation.
Collapse
Affiliation(s)
- Haruhiro Toko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan. .,Department of Advanced Translational Research and Medicine in Management of Pulmonary Hypertension, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masanori Katakura
- Department of Environmental Physiology, Faculty of Medicine, Shimane University, 89-1 Enyacho, Izumo, Shimane, 693-8501, Japan.,Laboratory of Nutritional Physiology, Department of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado, Saitama, 350-0295, Japan
| | - Michio Hashimoto
- Department of Environmental Physiology, Faculty of Medicine, Shimane University, 89-1 Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Toshiyuki Ko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Satoshi Bujo
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yusuke Adachi
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazutaka Ueda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Haruka Murakami
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masato Ishizuka
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Jiaxi Guo
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Chunxia Zhao
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takayuki Fujiwara
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hironori Hara
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Norifumi Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Osamu Shido
- Department of Environmental Physiology, Faculty of Medicine, Shimane University, 89-1 Enyacho, Izumo, Shimane, 693-8501, Japan
| | - Mutsuo Harada
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Department of Advanced Clinical Science and Therapeutics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|
13
|
Resolution-Associated Lactoferrin Peptides Limit LPS Signaling and Cytokine Secretion from Human Macrophages. Int J Mol Sci 2020; 21:ijms21145166. [PMID: 32708308 PMCID: PMC7404327 DOI: 10.3390/ijms21145166] [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: 06/12/2020] [Revised: 07/18/2020] [Accepted: 07/19/2020] [Indexed: 01/01/2023] Open
Abstract
The neutrophil granule protein lactoferrin is cleaved and accumulates in efferocytic macrophages as inflammation is resolved. Two peptides present within a resolution-associated 17 kDa fragment of lactoferrin promote the termination of inflammation in vivo by enhancing murine macrophage reprogramming. Here, we report that these two bioactive tripeptides, phenylalanine-lysine-aspartic acid and phenylalanine-lysine-glutamic acid (FKD and FKE, respectively), inhibit ERK and cJun activation following human macrophage exposure to LPS. In addition, these peptides at low concentrations (1–10 μM) modulate human macrophage reprogramming to an anti-inflammatory/pro-resolving phenotype. This was reflected by inhibition of LPS-induced TNF-α and IL-6 secretion and increased IL-10 levels. Moreover, we found naturally occurring FKE analogs (FKECH and FKECHLA) can recapitulate the activity of the short peptide in regulating macrophage cytokine secretion, whereas a reversed EKF peptide was inert in this respect. Curiously, FKD and FKE also regulated cytokine production by bone marrow-derived mouse macrophages, but in a very different fashion than their effect on human macrophages. Thus, lactoferrin peptides limit pro-inflammatory signaling and cytokine production by LPS-activated human macrophages and thereby enhance the resolution of inflammation.
Collapse
|
14
|
Avanoǧlu Güler A, Rossi FW, Bellando-Randone S, Prevete N, Tufan A, Manetti M, de Paulis A, Matucci-Cerinic M. The Role of Endogenous Eicosapentaenoic Acid and Docosahexaenoic Acid-Derived Resolvins in Systemic Sclerosis. Front Immunol 2020; 11:1249. [PMID: 32636845 PMCID: PMC7318896 DOI: 10.3389/fimmu.2020.01249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/18/2020] [Indexed: 12/19/2022] Open
Abstract
Resolvins, the member of specialized pro-resolving mediators, are produced from omega-3 polyunsaturated fatty acids as a response to an acute inflammatory process in that termination and resolution of inflammation. In the acute inflammation, these lipid mediators limit polymorphonuclear cells infiltration, proinflammatory cytokine production; promote efferocytosis, and regulate several cell types being important roles in innate and adaptive immunity. Any dysregulation or defect of the resolution phase result in prolonged, persistent inflammation and eventually fibrosis. Resolvins are implicated in the development of various chronic autoimmune diseases. Systemic sclerosis (SSc) is a very complicated, chronic autoimmune disorder proceeding with vasculopathy, inflammation, and fibrosis. Dysregulation of innate and adaptive immunity is another important contributing factor in the pathogenesis of SSc. In this review, we will focus on the different roles of this new family of lipid mediators, characterized by the ability to prevent the spread of inflammation and its chronicity in various ways and how they can control the development of fibrotic diseases like SSc.
Collapse
Affiliation(s)
- Aslıhan Avanoǧlu Güler
- Department of Experimental and Clinical Medicine, University of Florence and Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
- Department of Internal Medicine, Division of Rheumatology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Francesca Wanda Rossi
- Department of Internal Medicine, Clinical Immunology and Rheumatology, University of Naples Federico II, Naples, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, University of Florence and Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
| | - Nella Prevete
- Department of Internal Medicine, Clinical Immunology and Rheumatology, University of Naples Federico II, Naples, Italy
| | - Abdurrahman Tufan
- Department of Internal Medicine, Division of Rheumatology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, University of Florence and Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
| | - Amato de Paulis
- Department of Internal Medicine, Clinical Immunology and Rheumatology, University of Naples Federico II, Naples, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence and Department of Geriatric Medicine, Division of Rheumatology AOUC, Florence, Italy
| |
Collapse
|
15
|
Yaseen H, Butenko S, Polishuk-Zotkin I, Schif-Zuck S, Pérez-Sáez JM, Rabinovich GA, Ariel A. Galectin-1 Facilitates Macrophage Reprogramming and Resolution of Inflammation Through IFN-β. Front Pharmacol 2020; 11:901. [PMID: 32625094 PMCID: PMC7311768 DOI: 10.3389/fphar.2020.00901] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 06/02/2020] [Indexed: 12/15/2022] Open
Abstract
During the resolution of acute inflammation, macrophages undergo reprogramming from pro-inflammatory, to anti-inflammatory/reparative, and eventually to pro-resolving macrophages. Galectin-1 (Gal-1) is a bona fide pro-resolving lectin while interferon β (IFN-β) was recently shown to facilitate macrophage reprogramming and resolution of inflammation. In this study, we found Gal-1null mice exhibit a hyperinflammatory phenotype during the resolution of zymosan A-induced peritonitis but not during the early inflammatory response. This phenotype was characterized by reduced macrophage numbers, increased secretion of pro-inflammatory cytokines, such as interleukin-12 (IL-12), and reduced secretion of anti-inflammatory cytokines, such as interleukin-10 (IL-10). In addition, we found a delayed expression of the pro-resolving enzyme 12/15-lipoxygenase in macrophages and heightened levels of the inflammatory protease proteinase-3 (PR3) in peritoneal fluids from Gal-1null mice. Moreover, we observed sex-dependent differences in the inflammatory profile of Gal-1null mice. Notably, we found that IFN-β levels were reduced in resolution-phase exudates from Gal-1null mice. Administration of IFN-β in vivo or ex vivo treatment was able to rescue, at least in part, the hyperinflammatory profile of Gal-1null mice. In particular, IFN-β recovered a subset of F4/80+GR-1+ macrophages, restored IL-12 and IL-10 secretion from macrophages to WT values and diminished abnormal peritoneal PR3 levels in Gal-1null mice. In conclusion, our results revealed a new Gal-1-IFN-β axis that facilitates the resolution of inflammation and might restrain uncontrolled inflammatory disorders.
Collapse
Affiliation(s)
- Hiba Yaseen
- Departments of Biology and Human Biology, University of Haifa, Haifa, Israel
| | - Sergei Butenko
- Departments of Biology and Human Biology, University of Haifa, Haifa, Israel
| | | | - Sagie Schif-Zuck
- Departments of Biology and Human Biology, University of Haifa, Haifa, Israel
| | - Juan Manuel Pérez-Sáez
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Gabriel Adrian Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina.,Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Amiram Ariel
- Departments of Biology and Human Biology, University of Haifa, Haifa, Israel
| |
Collapse
|
16
|
Coppock GM, Aronson LR, Park J, Qiu C, Park J, DeLong JH, Radaelli E, Suszták K, Hunter CA. Loss of IL-27Rα Results in Enhanced Tubulointerstitial Fibrosis Associated with Elevated Th17 Responses. THE JOURNAL OF IMMUNOLOGY 2020; 205:377-386. [PMID: 32522836 DOI: 10.4049/jimmunol.1901463] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 05/11/2020] [Indexed: 12/15/2022]
Abstract
Clinical and experimental studies have established that immune cells such as alternatively activated (M2) macrophages and Th17 cells play a role in the progression of chronic kidney disease, but the endogenous pathways that limit these processes are not well understood. The cytokine IL-27 has been shown to limit immune-mediated pathology in other systems by effects on these cell types, but this has not been thoroughly investigated in the kidney. Unilateral ureteral obstruction was performed on wild-type and IL-27Rα-/- mice. After 2 wk, kidneys were extracted, and the degree of injury was measured by hydroxyproline assay and quantification of neutrophil gelatinase-associated lipocalin mRNA. Immune cell infiltrate was evaluated by immunohistochemistry and flow cytometry. An anti-IL-17A mAb was subsequently administered to IL-27Rα-/- mice every 2 d from day of surgery with evaluation as described after 2 wk. After unilateral ureteral obstruction, IL-27 deficiency resulted in increased tissue injury and collagen deposition associated with higher levels of chemokine mRNA and increased numbers of M2 macrophages. Loss of the IL-27Rα led to increased infiltration of activated CD4+ T cells that coproduced IL-17A and TNF-α, and blockade of IL-17A partially ameliorated kidney injury. Patients with chronic kidney disease had elevated serum levels of IL-27 and IL-17A, whereas expression of transcripts for the IL-27RA and the IL-17RA in the tubular epithelial cells of patients with renal fibrosis correlated with disease severity. These data suggest that endogenous IL-27 acts at several points in the inflammatory cascade to limit the magnitude of immune-mediated damage to the kidney.
Collapse
Affiliation(s)
- Gaia M Coppock
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104.,Renal, Electrolyte, and Hypertension Division, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Lillian R Aronson
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104.,Section of Surgery, Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jihwan Park
- Renal, Electrolyte, and Hypertension Division, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Chengxiang Qiu
- Renal, Electrolyte, and Hypertension Division, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Jeongho Park
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Jonathan H DeLong
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Enrico Radaelli
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Katalin Suszták
- Renal, Electrolyte, and Hypertension Division, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Christopher A Hunter
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104;
| |
Collapse
|
17
|
Characterization of Chenopodin Isoforms from Quinoa Seeds and Assessment of Their Potential Anti-Inflammatory Activity in Caco-2 Cells. Biomolecules 2020; 10:biom10050795. [PMID: 32455586 PMCID: PMC7277664 DOI: 10.3390/biom10050795] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/12/2020] [Accepted: 05/18/2020] [Indexed: 01/18/2023] Open
Abstract
Several food-derived molecules, including proteins and peptides, can show bioactivities toward the promotion of well-being and disease prevention in humans. There is still a lack of information about the potential effects on immune and inflammatory responses in mammalian cells following the ingestion of seed storage proteins. This study, for the first time, describes the potential immunomodulation capacity of chenopodin, the major protein component of quinoa seeds. After characterizing the molecular features of the purified protein, we were able to separate two different forms of chenopodin, indicated as LcC (Low charge Chenopodin, 30% of total chenopodin) and HcC (High charge Chenopodin, 70% of total chenopodin). The biological effects of LcC and HcC were investigated by measuring NF-κB activation and IL-8 expression studies in undifferentiated Caco-2 cells. Inflammation was elicited using IL-1β. The results indicate that LcC and HcC show potential anti-inflammatory activities in an intestinal cell model, and that the proteins can act differently, depending on their structural features. Furthermore, the molecular mechanisms of action and the structural/functional relationships of the protein at the basis of the observed bioactivity were investigated using in silico analyses and structural predictions.
Collapse
|
18
|
Prawira AY, Hosaka YZ, Novelina S, Farida WR, Darusman HS, Agungpriyono S. Morphological evaluation of polysaccharide content and collagen composition during cutaneous wound healing in the Sunda porcupine (Hystrix javanica). J Vet Med Sci 2020; 82:506-515. [PMID: 32213731 PMCID: PMC7273595 DOI: 10.1292/jvms.19-0603] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Wound healing in the Sunda porcupine is believed to occur quickly, although the wound is large and severe. Wound enclosure involves many processes to restore the lost or damaged skin
structure where conjugated polysaccharide-protein and collagen, as the main components deposited in wound tissue to restore it. The aim of this study was to evaluate alteration of
polysaccharide contents and collagen in untreated full-thickness wound healing in the thoracodorsal and lumbosacral regions in the Sunda porcupines. Histological analysis was performed by
periodic acid Schiff, alcian blue pH 2.5, picrosirius red staining method and Low Vacuum Scanning Electron Microscope (LV-SEM) imaging to obtain the fundamental data of healing process.
Wound healing began with re-epithelization followed by progressive wound contraction with 4 overlapping stages in about 30–50 days until the wound closed (21–30 days in thoracodorsal and
30–50 days in lumbosacral). Neutral polysaccharide was more widely distributed compared to the acid polysaccharide in almost all stages of wound healing. The ratio of collagen I to III
appeared to be higher in the thoracodorsal region than the lumbosacral region during healing process. LV-SEM imaging showed changes in connective tissue structure in the wound border and
granulation tissue which appeared abundant and mixed of thin and thick fiber. In conclusion, cutaneous full thickness wound healing in the Sunda porcupine occurred faster in the
thoracodorsal region, which might be correlated to the role of neutral polysaccharide and a high ratio of collagen I to III.
Collapse
Affiliation(s)
- Andhika Yudha Prawira
- Department of Anatomy Physiology and Pharmacology, Faculty of Veterinary Medicine, IPB University, Bogor 16680, Indonesia
| | - Yoshinao Z Hosaka
- Laboratory of Veterinary Anatomy, Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, Tottori 680-8550, Japan
| | - Savitri Novelina
- Department of Anatomy Physiology and Pharmacology, Faculty of Veterinary Medicine, IPB University, Bogor 16680, Indonesia
| | - Wartika Rosa Farida
- Zoology Division, Research Center for Biology, Indonesian Institute of Sciences, Cibinong 16911, Indonesia
| | - Huda Shalahudin Darusman
- Department of Anatomy Physiology and Pharmacology, Faculty of Veterinary Medicine, IPB University, Bogor 16680, Indonesia.,Primate Research Center, IPB University, Bogor 16680, Indonesia
| | - Srihadi Agungpriyono
- Department of Anatomy Physiology and Pharmacology, Faculty of Veterinary Medicine, IPB University, Bogor 16680, Indonesia
| |
Collapse
|
19
|
Vadivel S, Vincent P, Sekaran S, Visaga Ambi S, Muralidar S, Selvaraj V, Palaniappan B, Thirumalai D. Inflammation in myocardial injury- Stem cells as potential immunomodulators for myocardial regeneration and restoration. Life Sci 2020; 250:117582. [PMID: 32222465 DOI: 10.1016/j.lfs.2020.117582] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 03/14/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022]
Abstract
The ineffective immunosuppressant's and targeted strategies to neutralize inflammatory mediators have worsened the scenario of heart failure and have opened many questions for debate. Stem cell therapy has proven to be a promising approach for treating heart following myocardial infarction (MI). Adult stem cells, induced pluripotent stem cells and embryonic stem cells are possible cell types and have successfully shown to regenerate damaged myocardial tissue in pre-clinical and clinical studies. Current implications of using mesenchymal stem cells (MSCs) owing to their immunomodulatory functions and paracrine effects could serve as an effective alternative treatment option for rejuvenating the heart post MI. The major setback associated with the use of MSCs is reduced cell retention, engraftment and decreased effectiveness. With a few reports on understanding the role of inflammation and its dual effects on the structure and function of heart, this review focuses on these missing insights and further exemplifies the role of MSCs as an alternative therapy in treating the pathological consequences in myocardial infarction (MI).
Collapse
Affiliation(s)
- Sajini Vadivel
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Preethi Vincent
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Saravanan Sekaran
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India.
| | - Senthil Visaga Ambi
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India.
| | - Shibi Muralidar
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Vimalraj Selvaraj
- Centre for Biotechnology, Anna University, Chennai 600 025, Tamil Nadu, India
| | - Balamurugan Palaniappan
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| | - Diraviyam Thirumalai
- School of Chemical and Biotechnology, SASTRA Deemed-to-be-University, Thanjavur 613 401, Tamil Nadu, India
| |
Collapse
|
20
|
Viola H, Chang J, Grunwell JR, Hecker L, Tirouvanziam R, Grotberg JB, Takayama S. Microphysiological systems modeling acute respiratory distress syndrome that capture mechanical force-induced injury-inflammation-repair. APL Bioeng 2019; 3:041503. [PMID: 31768486 PMCID: PMC6874511 DOI: 10.1063/1.5111549] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 11/08/2019] [Indexed: 12/14/2022] Open
Abstract
Complex in vitro models of the tissue microenvironment, termed microphysiological systems, have enormous potential to transform the process of discovering drugs and disease mechanisms. Such a paradigm shift is urgently needed in acute respiratory distress syndrome (ARDS), an acute lung condition with no successful therapies and a 40% mortality rate. Here, we consider how microphysiological systems could improve understanding of biological mechanisms driving ARDS and ultimately improve the success of therapies in clinical trials. We first discuss how microphysiological systems could explain the biological mechanisms underlying the segregation of ARDS patients into two clinically distinct phenotypes. Then, we contend that ARDS-mimetic microphysiological systems should recapitulate three critical aspects of the distal airway microenvironment, namely, mechanical force, inflammation, and fibrosis, and we review models that incorporate each of these aspects. Finally, we recognize the substantial challenges associated with combining inflammation, fibrosis, and/or mechanical force in microphysiological systems. Nevertheless, complex in vitro models are a novel paradigm for studying ARDS, and they could ultimately improve patient care.
Collapse
Affiliation(s)
| | - Jonathan Chang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia 30332, USA
| | - Jocelyn R. Grunwell
- Department of Pediatrics, Division of Critical Care Medicine, Children's Healthcare of Atlanta at Egleston, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Louise Hecker
- Division of Pulmonary, Allergy and Critical Care and Sleep Medicine, University of Arizona, Tucson, Arizona 85724, USA and Southern Arizona Veterans Affairs Health Care System, Tucson, Arizona 85723, USA
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia 30322, USA and Center for CF and Airways Disease Research, Children's Healthcare of Atlanta, Atlanta, Georgia 30322, USA
| | - James B. Grotberg
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | |
Collapse
|
21
|
Carillon J, Saby M, Barial S, Sansone A, Scanferlato R, Gayrard N, Lajoix AD, Jover B, Chatgilialoglu C, Ferreri C. Melon juice concentrate supplementation in an animal model of obesity: Involvement of relaxin and fatty acid pathways. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
22
|
Abstract
Tissue-resident macrophages shape the development and homeostasis of the organs that they colonize. Yu et al. (2017) demonstrate that, unique to the lung, the instructive cytokine TGF-β collaborates with GM-CSF to induce the master transcription factor PPARγ to drive alveolar macrophage differentiation and maintenance early in life.
Collapse
|
23
|
The MicroRNA miR-155 Is Essential in Fibrosis. Noncoding RNA 2019; 5:ncrna5010023. [PMID: 30871125 PMCID: PMC6468348 DOI: 10.3390/ncrna5010023] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/04/2019] [Accepted: 03/07/2019] [Indexed: 02/07/2023] Open
Abstract
The function of microRNAs (miRNAs) during fibrosis and the downstream regulation of gene expression by these miRNAs have become of great biological interest. miR-155 is consistently upregulated in fibrotic disorders, and its ablation downregulates collagen synthesis. Studies demonstrate the integral role of miR-155 in fibrosis, as it mediates TGF-β1 signaling to drive collagen synthesis. In this review, we summarize recent findings on the association between miR-155 and fibrotic disorders. We discuss the cross-signaling between macrophages and fibroblasts that orchestrates the upregulation of collagen synthesis mediated by miR-155. As miR-155 is involved in the activation of the innate and adaptive immune systems, specific targeting of miR-155 in pathologic cells that make excessive collagen could be a viable option before the depletion of miR-155 becomes an attractive antifibrotic approach.
Collapse
|
24
|
Kourtzelis I, Li X, Mitroulis I, Grosser D, Kajikawa T, Wang B, Grzybek M, von Renesse J, Czogalla A, Troullinaki M, Ferreira A, Doreth C, Ruppova K, Chen LS, Hosur K, Lim JH, Chung KJ, Grossklaus S, Tausche AK, Joosten LA, Moutsopoulos NM, Wielockx B, Castrillo A, Korostoff JM, Coskun Ü, Hajishengallis G, Chavakis T. DEL-1 promotes macrophage efferocytosis and clearance of inflammation. Nat Immunol 2019; 20:40-49. [PMID: 30455459 PMCID: PMC6291356 DOI: 10.1038/s41590-018-0249-1] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022]
Abstract
Resolution of inflammation is essential for tissue homeostasis and represents a promising approach to inflammatory disorders. Here we found that developmental endothelial locus-1 (DEL-1), a secreted protein that inhibits leukocyte-endothelial adhesion and inflammation initiation, also functions as a non-redundant downstream effector in inflammation clearance. In human and mouse periodontitis, waning of inflammation was correlated with DEL-1 upregulation, whereas resolution of experimental periodontitis failed in DEL-1 deficiency. This concept was mechanistically substantiated in acute monosodium-urate-crystal-induced inflammation, where the pro-resolution function of DEL-1 was attributed to effective apoptotic neutrophil clearance (efferocytosis). DEL-1-mediated efferocytosis induced liver X receptor-dependent macrophage reprogramming to a pro-resolving phenotype and was required for optimal production of at least certain specific pro-resolving mediators. Experiments in transgenic mice with cell-specific overexpression of DEL-1 linked its anti-leukocyte-recruitment action to endothelial cell-derived DEL-1 and its efferocytic/pro-resolving action to macrophage-derived DEL-1. Thus, the compartmentalized expression of DEL-1 facilitates distinct homeostatic functions in an appropriate context that can be harnessed therapeutically.
Collapse
Affiliation(s)
- Ioannis Kourtzelis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Xiaofei Li
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis Mitroulis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Daniel Grosser
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Tetsuhiro Kajikawa
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Baomei Wang
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michal Grzybek
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Janusz von Renesse
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Aleksander Czogalla
- Laboratory of Cytobiochemistry, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Maria Troullinaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Anaisa Ferreira
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christian Doreth
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Klara Ruppova
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Lan-Sun Chen
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Kavita Hosur
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jong-Hyung Lim
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Kyoung-Jin Chung
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Sylvia Grossklaus
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Anne Kathrin Tausche
- Division of Rheumatology, Medical Clinic III, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Leo A.B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Ben Wielockx
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Antonio Castrillo
- Instituto de Investigaciones Biomedicas “Alberto Sols” Consejo Superior de Investigaciones Científicas (CSIC) de Madrid, Spain
- Unidad de Biomedicina IIBM-ULPGC (Unidad Asociada al CSIC), Instituto Universitario de Investigaciones Biomedicas y Sanitarias (IUIBS) de la ULPGC, Las Palmas, Spain
| | - Jonathan M. Korostoff
- Department of Periodontics, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ünal Coskun
- Paul Langerhans Institute Dresden of the Helmholtz Zentrum München at the University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - George Hajishengallis
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
25
|
Thieblemont N, Witko-Sarsat V, Ariel A. Regulation of macrophage activation by proteins expressed on apoptotic neutrophils: Subversion towards autoimmunity by proteinase 3. Eur J Clin Invest 2018; 48 Suppl 2:e12990. [PMID: 30039869 DOI: 10.1111/eci.12990] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/27/2018] [Indexed: 12/13/2022]
Abstract
Neutrophils are critically involved in host defence and they also modulate the inflammatory process. Turning the inflammatory response towards a resolutive outcome requires a dialogue between apoptotic neutrophils and proresolving macrophages through complex key molecular interactions controlling efferocytosis, anti-inflammatory reprogramming and ultimately immune regulation. In this review, we will first focus on recent molecular analyses aiming at characterizing the role of proteins expressed on apoptotic neutrophils and their cognate partners expressed on macrophages in the resolution of inflammation. These will include chemokine receptors and their ligands and annexin A1 and its receptor FPR2. We will next depict how the structural and enzymatic properties of proteinase 3 (PR3), the autoantigen in vasculitis, allow its expression on apoptotic neutrophils, which in turn affects efferocytosis and immune response associated with the clearance of apoptotic cells. This example illustrates that the fate of apoptotic neutrophils directly influences the resolution of inflammation and immune responses thereby potentially contributing to systemic and nonresolving inflammation as well as autoimmunity.
Collapse
Affiliation(s)
- Nathalie Thieblemont
- INSERM U1016 Cochin Institute, Paris, France.,CNRS UMR 8104, Paris, France.,Université Paris-Descartes, Paris, France.,Center of Excellence LABEX Inflamex, Paris, France
| | - Véronique Witko-Sarsat
- INSERM U1016 Cochin Institute, Paris, France.,CNRS UMR 8104, Paris, France.,Université Paris-Descartes, Paris, France.,Center of Excellence LABEX Inflamex, Paris, France
| | - Amiram Ariel
- Department of Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
26
|
Lu S, Herzlinger M, Cao W, Noble L, Yang D, Shapiro J, Kurtis J, LeLeiko N, Resnick M. Utility of 15(S)-HETE as a Serological Marker for Eosinophilic Esophagitis. Sci Rep 2018; 8:14498. [PMID: 30266946 PMCID: PMC6162315 DOI: 10.1038/s41598-018-32944-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 09/19/2018] [Indexed: 12/30/2022] Open
Abstract
The pathogenesis of eosinophilic esophagitis (EoE) involves Th2-mediated eosinophil recruitment and degranulation into the esophagus. However, measuring serum Th2 cytokines, eosinophils, and eosinophil-derived products does not reliably distinguish EoE from control populations. Non-invasive methods to diagnose EoE are lacking. We evaluated the diagnostic value of a novel candidate biomarker of EoE: 15(S)-hydroxyeicosatetraenoic acid (HETE). We used immunoassay to measure 15(S)-HETE and cytokine profiles in patients undergoing endoscopy with known or suspected EoE. 31 subjects were enrolled, 16 with EoE, and 15 with an alternate diagnosis. 15(S)-HETE was elevated in the EoE group compared to non-EoE group. The sensitivity and specificity of 15(S)-HETE to be used as a non-invasive marker is 50% and 80%, respectively. 15(S)-HETE may aid in the diagnosis of EoE.
Collapse
Affiliation(s)
- Shaolei Lu
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, USA.
| | - Michael Herzlinger
- Division of Pediatric Gastroenterology, Nutrition, and Liver Diseases, Hasbro Children's Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| | - Weibiao Cao
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| | - Lelia Noble
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| | - Dongfang Yang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| | - Jason Shapiro
- Division of Pediatric Gastroenterology, Nutrition, and Liver Diseases, Hasbro Children's Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| | - Jonathan Kurtis
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| | - Neal LeLeiko
- Division of Pediatric Gastroenterology, Nutrition, and Liver Diseases, Hasbro Children's Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| | - Murray Resnick
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, USA
| |
Collapse
|
27
|
Sanguigno L, Casamassa A, Funel N, Minale M, Riccio R, Riccio S, Boscia F, Brancaccio P, Pollina LE, Anzilotti S, Di Renzo G, Cuomo O. Triticum vulgare extract exerts an anti-inflammatory action in two in vitro models of inflammation in microglial cells. PLoS One 2018; 13:e0197493. [PMID: 29902182 PMCID: PMC6002026 DOI: 10.1371/journal.pone.0197493] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/03/2018] [Indexed: 01/08/2023] Open
Abstract
Triticum vulgare has been extensively used in traditional medicine thanks to its properties of accelerating tissue repair. The specific extract of Triticum vulgare manufactured by Farmaceutici Damor (TVE-DAMOR) is already present in some pharmaceutical formulations used in the treatment of decubitus ulcers, skin lesions and burns. It has been recently suggested that this Triticum vulgare extract may possess potential anti-inflammatory properties. In the light of these premises the aim of the present paper was to verify the anti-inflammatory role of TVE, using the LPS-stimulated microglia model of inflammation. In particular the effect of different concentrations of TVE on the release of several mediators of inflammation such as nitric oxide, IL-6, PGE2 and TNF alpha was evaluated. More important, the anti-inflammatory effect of TVE was confirmed also in primary rat microglia cultures. The results of the present study show that TVE exerts anti-inflammatory properties since it reduces the release of all the evaluated markers of inflammation, such as NO, IL6, TNF alpha and PGE2 in LPS-activated BV2 microglial cells. Intriguingly, TVE reduced microglia activation and NO release also in primary microglia. Indeed, to verify the pathway of modulation of the inflammatory markers reported above, we found that TVE restores the cytoplasmic expression of p65 protein, kwown as specific marker associated with activation of inflammatory response. The evidence for an inhibitory activity on inflammation of this specific extract of Triticum vulgare may open the way to the possibility of a therapeutical use of the Triticum vulgare extract as an anti-inflammatory compound in certain pathological states such as burns, decubitus ulcers, folliculitis and inflammation of peripheral nerve.
Collapse
Affiliation(s)
- Luca Sanguigno
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Antonella Casamassa
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Niccola Funel
- Department of Translational Research and The New Technologies in Medicine and Surgery, University of Pisa, Italy
| | | | | | | | - Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Paola Brancaccio
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | | | | | - Gianfranco Di Renzo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Ornella Cuomo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| |
Collapse
|
28
|
Subiela JV, Torres SH, De Sanctis JB, Hernández N. Cardiorespiratory responses, nitric oxide production and inflammatory factors in patients with myocardial infarction after rehabilitation. Nitric Oxide 2018. [DOI: 10.1016/j.niox.2018.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
29
|
Kuba A, Raida L. Graft versus Host Disease: From Basic Pathogenic Principles to DNA Damage Response and Cellular Senescence. Mediators Inflamm 2018; 2018:9451950. [PMID: 29785172 PMCID: PMC5896258 DOI: 10.1155/2018/9451950] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 02/12/2018] [Accepted: 02/21/2018] [Indexed: 12/14/2022] Open
Abstract
Graft versus host disease (GVHD), a severe immunogenic complication of allogeneic hematopoietic stem cell transplantation (HSCT), represents the most frequent cause of transplant-related mortality (TRM). Despite a huge progress in HSCT techniques and posttransplant care, GVHD remains a significant obstacle in successful HSCT outcome. This review presents a complex summary of GVHD pathogenesis with focus on references considering basic biological processes such as DNA damage response and cellular senescence.
Collapse
Affiliation(s)
- Adam Kuba
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Ludek Raida
- Department of Hemato-Oncology, University Hospital and Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
30
|
Millet P, Opiekun M, Martin T, Beauchamp GK, Kimball BA. Cytokine contributions to alterations of the volatile metabolome induced by inflammation. Brain Behav Immun 2018; 69:312-320. [PMID: 29241669 DOI: 10.1016/j.bbi.2017.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/28/2017] [Accepted: 12/09/2017] [Indexed: 01/02/2023] Open
Abstract
Several studies demonstrate that inflammation affects body odor. Volatile signals associated with inflammation induced by pyrogens like LPS are detectable both by conspecifics and chemical analyses. However, little is known about the mechanisms which translate detection of a foreign molecule or pathogen into a unique body odor, or even how unique that odor may be. Here, we utilized C57BL/6J trained mice to identify the odor of LPS-treated conspecifics to investigate potential pathways between LPS-induced inflammation and changes in body odor, as represented by changes in urine odor. We hypothesized that the change in volatile metabolites could be caused directly by the pro-inflammatory cytokine response mediated by TNF or IL-1β, or by the compensatory anti-inflammatory response mediated by IL-10. We found that trained biosensors generalized learned LPS-associated odors to TNF-induced odors, but not to IL-1β or IL-10-induced odors. Analyses of urine volatiles using headspace gas chromatography revealed distinct profiles of volatile compounds for each treatment. Instrumental discrimination relied on a mixture of compounds, including 2-sec-butyl-4,5-dihydrothiazole, cedrol, nonanal, benzaldehyde, acetic acid, 2-ethyl-1-hexanol, and dehydro-exo-brevicomin. Although interpretation of LDA modeling differed from behavioral testing, it does suggest that treatment with TNF, IL-1β, and LPS can be distinguished by their resultant volatile profiles. These findings indicate there is information found in body odors on the presence of specific cytokines. This result is encouraging for the future of disease diagnosis via analysis of volatiles.
Collapse
Affiliation(s)
- Patrick Millet
- Monell Chemical Senses Center, Philadelphia, PA, United States.
| | | | - Talia Martin
- Monell Chemical Senses Center, Philadelphia, PA, United States
| | | | - Bruce A Kimball
- USDA-APHIS-WS National Wildlife Research Center, Monell Chemical Senses Center, Philadelphia, PA, United States
| |
Collapse
|
31
|
Esendagli G, Yoyen-Ermis D, Guseinov E, Aras C, Aydin C, Uner A, Hamaloglu E, Karakoc D. Impact of repeated abdominal surgery on wound healing and myeloid cell dynamics. J Surg Res 2018; 223:188-197. [PMID: 29433873 DOI: 10.1016/j.jss.2017.11.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/13/2017] [Accepted: 11/10/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Even though wound dehiscence is a surgical complication, under certain medical conditions, repetition of the laparotomy (LT) (relaparotomy) can become inevitable. In addition to the risks associated with this surgical operation, relaparotomy can interfere with the tissue healing and contribute to the development of chronic wounds. METHODS In an experimental relaparotomy wounding model, this study investigated the impact of repeated surgery on wound healing and on the immune cells of myeloid origin. RESULTS The first repeat of the LT triggered fibrosis and marginally interfered with the wound healing; however, the second operation completely abrogated the healing process. Splenomegaly was observed as an indicator of the chronic inflammation and the systemic effect of repeated laparotomies. In the blood stream, the spleen, and the liver, these repeated surgeries exhibited a major impact on the CD11b+Ly6C+Ly6G- monocytes. On the other hand, especially, whespecially the second relaparotomy resulted in a massive purging of neutrophil granulocytes into the circulation. These CD11b+Ly6C+Ly6G+ neutrophils that were disseminated on repeated abdominal laparotomies had a proinflammatory character that positively influenced T cell proliferation and displayed a high capacity for production of reactive oxygen species. CONCLUSIONS The repetition of abdominal LT not only interferes with the wound healing but also contributes to the development of imperfectly healing wounds which have systemic impact on immune compartments.
Collapse
Affiliation(s)
- Gunes Esendagli
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey.
| | - Digdem Yoyen-Ermis
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Emil Guseinov
- Department of General Surgery, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Cigdem Aras
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Cisel Aydin
- Department of Pathology, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Aysegul Uner
- Department of Pathology, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Erhan Hamaloglu
- Department of General Surgery, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Derya Karakoc
- Department of General Surgery, Hacettepe University Medical Faculty, Ankara, Turkey
| |
Collapse
|
32
|
Cerveró-Ferragut S, López-Riquelme N, Martín-Tomás E, Massa-Domínguez B, Pomares-Vicente J, Soler-Pérez M, Sánchez-Hernández JF. Quantitative analysis of blood cells and inflammatory factors in wounds. J Wound Care 2017; 26:121-125. [PMID: 28277999 DOI: 10.12968/jowc.2017.26.3.121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The aim of this study was to quantify blood cells and inflammatory markers, involved in the healing process, in exudates from wounds in different healing phases, to assess these markers in order to identify the inflammatory phase of the wounds. METHOD Patients who presented with postsurgical wounds, which closed by first and second intention, and those who presented with pressure ulcers (PUs), which were closed by second intention, were included in the study. RESULTS We examined wounds from 37 patients and collected samples from 52 wounds in the inflammatory phase, 30 in the proliferative phase and 29 in the maturation phase. The number of neutrophils and platelets in the exudate collected from wounds in the inflammatory phase was significantly higher (p<0.001), while the number of lymphocytes, was significantly lower in exudate from wounds in the inflammatory phase (p<0.001). Wound c-reactive protein (CRP) and immunoglobulin G (IgG) levels were higher in the inflammatory group (p<0.001). We found a significantly positive correlation between CRP levels and the percentage of neutrophils and monocytes (r=0.346, p=0.004; r=0.293, p=0.015), and a significantly negative correlation between CRP levels and the percentage of lymphocytes (r=-0.503, p<0.001). A stepwise logistic regression analysis was used to identify an optimal combination of these biomarkers. The optimal biomarker combinations were neutrophils + monocytes + platelets + IgG + CRP, with an area under the curve (AUC) of 0.981 [confidence interval (CI) 95%: 0.955-1.000, p<0.001] for the diagnosis of wounds in the inflammatory phase. The optimal cutpoint yielded 96.9 % sensitivity and 94.6 % specificity. The biomarker combination predicted the inflammatory phase and was superior to individual biomarkers. CONCLUSION Our findings suggest that the combination of the markers, percentage of neutrophils and monocytes, platelets, CRP and IgG levels could be useful prognostic indicators of the inflammatory phase.
Collapse
Affiliation(s)
- S Cerveró-Ferragut
- Wound Department, General University Hospital of Elche (Alicante), Spain
| | - N López-Riquelme
- Department of Laboratory Medicine, University Clinical Hospital Virgen de la Arrixaca, El Palmar (Murcia), Spain
| | - E Martín-Tomás
- Laboratory Medicine Department, General University Hospital of Elche (Alicante), Spain
| | - B Massa-Domínguez
- Wound Department, General University Hospital of Elche (Alicante), Spain
| | - J Pomares-Vicente
- Wound Department, General University Hospital of Elche (Alicante), Spain
| | - M Soler-Pérez
- Wound Department, General University Hospital of Elche (Alicante), Spain
| | - J F Sánchez-Hernández
- Laboratory Medicine Department, General University Hospital of Elche (Alicante), Spain
| |
Collapse
|
33
|
Xu C, Fu F, Li X, Zhang S. Mesenchymal stem cells maintain the microenvironment of central nervous system by regulating the polarization of macrophages/microglia after traumatic brain injury. Int J Neurosci 2017; 127:1124-1135. [PMID: 28464695 DOI: 10.1080/00207454.2017.1325884] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs), which are regarded as promising candidates for cell replacement therapies, are able to regulate immune responses after traumatic brain injury (TBI). Secondary immune response following the mechanical injury is the essential factor leading to the necrosis and apoptosis of neural cells during and after the cerebral edema has subsided and there is lack of efficient agent that can mitigate such neuroinflammation in the clinical application. By means of three molecular pathways (prostaglandin E2 (PGE2), tumor-necrosis-factor-inducible gene 6 protein (TSG-6), and progesterone receptor (PR) and glucocorticoid receptors (GR)), MSCs induce the activation of macrophages/microglia and drive them polarize into the M2 phenotypes, which inhibits the release of pro-inflammatory cytokines and promotes tissue repair and nerve regeneration. The regulation of MSCs and the polarization of macrophages/microglia are dynamically changing based on the inflammatory environment. Under the stimulation of platelet lysate (PL), MSCs also promote the release of pro-inflammatory cytokines. Meanwhile, the statue of macrophages/microglia exerts significant effects on the survival, proliferation, differentiation and activation of MSCs by changing the niche of cells. They form positive feedback loops in maintaining the homeostasis after TBI to relieving the secondary injury and promoting tissue repair. MSC therapies have obtained great achievements in several central nervous system disease clinical trials, which will accelerate the application of MSCs in TBI treatment.
Collapse
Affiliation(s)
- Chao Xu
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| | - Feng Fu
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| | - Xiaohong Li
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| | - Sai Zhang
- a Institute of Traumatic Brain Injury and Neurology, Pingjin Hospital , Logistics University of Chinese People's Armed Police Forces , Tianjin 300162 , China
| |
Collapse
|
34
|
Cooke KR, Luznik L, Sarantopoulos S, Hakim FT, Jagasia M, Fowler DH, van den Brink MRM, Hansen JA, Parkman R, Miklos DB, Martin PJ, Paczesny S, Vogelsang G, Pavletic S, Ritz J, Schultz KR, Blazar BR. The Biology of Chronic Graft-versus-Host Disease: A Task Force Report from the National Institutes of Health Consensus Development Project on Criteria for Clinical Trials in Chronic Graft-versus-Host Disease. Biol Blood Marrow Transplant 2017; 23:211-234. [PMID: 27713092 PMCID: PMC6020045 DOI: 10.1016/j.bbmt.2016.09.023] [Citation(s) in RCA: 304] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
Abstract
Chronic graft-versus-host disease (GVHD) is the leading cause of late, nonrelapse mortality and disability in allogeneic hematopoietic cell transplantation recipients and a major obstacle to improving outcomes. The biology of chronic GVHD remains enigmatic, but understanding the underpinnings of the immunologic mechanisms responsible for the initiation and progression of disease is fundamental to developing effective prevention and treatment strategies. The goals of this task force review are as follows: This document is intended as a review of our understanding of chronic GVHD biology and therapies resulting from preclinical studies, and as a platform for developing innovative clinical strategies to prevent and treat chronic GVHD.
Collapse
Affiliation(s)
- Kenneth R Cooke
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland.
| | - Leo Luznik
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland
| | - Stefanie Sarantopoulos
- Division of Hematological Malignancies and Cellular Therapy, Department of Immunology and Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Frances T Hakim
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Madan Jagasia
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Daniel H Fowler
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Marcel R M van den Brink
- Departments of Immunology and Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John A Hansen
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington, Seattle, Washington
| | - Robertson Parkman
- Division of Pediatric Stem Cell Transplantation and Regenerative Medicine, Stanford University, Palo Alto, California
| | - David B Miklos
- Division of Blood and Marrow Transplantation, Stanford University, Palo Alto, California
| | - Paul J Martin
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington, Seattle, Washington
| | - Sophie Paczesny
- Departments of Pediatrics and Immunology, Wells Center for Pediatric Research, Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana
| | - Georgia Vogelsang
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins Hospital, Baltimore, Maryland
| | - Steven Pavletic
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kirk R Schultz
- Michael Cuccione Childhood Cancer Research Program, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Bruce R Blazar
- Masonic Cancer Center and Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, Minnesota.
| |
Collapse
|
35
|
Lin C, Rezaee F, Waasdorp M, Shi K, van der Poll T, Borensztajn K, Spek CA. Protease activated receptor-1 regulates macrophage-mediated cellular senescence: a risk for idiopathic pulmonary fibrosis. Oncotarget 2016; 6:35304-14. [PMID: 26474459 PMCID: PMC4742106 DOI: 10.18632/oncotarget.6095] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/24/2015] [Indexed: 12/30/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a destructive disease in part resulting from premature or mature cellular aging. Protease-activated receptor-1 (PAR-1) recently emerged as a critical component in the context of fibrotic lung diseases. Therefore, we aimed to study the role of macrophages in PAR-1-mediated idiopathic pulmonary fibrosis. The number of macrophages were significantly reduced in lungs of PAR-1 antagonist (P1pal-12) treated animals upon bleomycin instillation. In line with these data, PAR-1 stimulation increased monocyte/macrophage recruitment in response to epithelium injury in in vitro trans-well assays. Moreover, macrophages induced fibroblasts migration, differentiation and secretion of collagen, which were inhibited in the presence of TGF-β receptor inhibitors. Interestingly, these profibrotic effects were partially inhibited by treatment with the PAR-1 inhibitor P1pal-12. Using shRNA mediated PAR-1 knock down in fibroblasts, we demonstrate that fibroblast PAR-1 contributes to TGF-β activation and production. Finally, we show that the macrophage-dependent induction of PAR-1 driven TGF-β activation was mediated by FXa. Our data identify novel mechanisms by which PAR-1 stimulation on different cell types can contribute to IPF and identify macrophages as key players in PAR-1 dependent development of this devastating disease. IPF may result from cellular senescence mediated by macrophages in the lung.
Collapse
Affiliation(s)
- Cong Lin
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Farhad Rezaee
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands.,Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen,The Netherlands
| | - Maaike Waasdorp
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Kun Shi
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Keren Borensztajn
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands.,Inserm UMR1152, Medical School Xavier Bichat, Paris, France.,Département Hospitalo-universtaire FIRE and LabEx Inflamex, Paris, France
| | - C Arnold Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
36
|
Williams JP, Calvi L, Chakkalakal JV, Finkelstein JN, O’Banion MK, Puzas E. Addressing the Symptoms or Fixing the Problem? Developing Countermeasures against Normal Tissue Radiation Injury. Radiat Res 2016; 186:1-16. [PMID: 27332954 PMCID: PMC4991354 DOI: 10.1667/rr14473.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Laura Calvi
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Joe V. Chakkalakal
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Jacob N. Finkelstein
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York
| | - M. Kerry O’Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York
| | - Edward Puzas
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
37
|
Ariel A, Ravichandran KS. 'This way please': Apoptotic cells regulate phagocyte migration before and after engulfment. Eur J Immunol 2016; 46:1583-6. [PMID: 27345468 DOI: 10.1002/eji.201646505] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 05/23/2016] [Accepted: 05/25/2016] [Indexed: 12/13/2022]
Abstract
Apoptotic leukocyte clearance is a hallmark of the resolution of inflammation and is a central fate-determining event for macrophages. The directional migration of motile phagocytes toward cellular corpses and the subsequent engulfment are tightly regulated, and the exciting molecular mechanisms for these complex steps are actively under investigation. In this issue Angsana et al. [Eur. J. Immunol. 2016. 46: 1592-1599.] report that the chemokine receptor CXCR4 is upregulated on murine and human macrophages following the engulfment of apoptotic cells, or following exposure to the pro-resolving nucleotide adenosine. This work, together with other recent findings, point toward a new mode of regulation of macrophages following the engulfment of apoptotic cells. In this commentary, we put these findings in relevant perspective and highlight its potential ramifications.
Collapse
Affiliation(s)
- Amiram Ariel
- Departments of Biology and Human Biology, University of Haifa, Israel
| | - Kodi S Ravichandran
- Center for Cell Clearance, Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
38
|
Lin C, von der Thüsen J, Isermann B, Weiler H, van der Poll T, Borensztajn K, Spek CA. High endogenous activated protein C levels attenuates bleomycin-induced pulmonary fibrosis. J Cell Mol Med 2016; 20:2029-2035. [PMID: 27295971 PMCID: PMC5082406 DOI: 10.1111/jcmm.12891] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/02/2016] [Indexed: 01/13/2023] Open
Abstract
Coagulation activation accompanied by reduced anticoagulant activity is a key characteristic of patients with idiopathic pulmonary fibrosis (IPF). Although the importance of coagulation activation in IPF is well studied, the potential relevance of endogenous anticoagulant activity in IPF progression remains elusive. We assess the importance of the endogenous anticoagulant protein C pathway on disease progression during bleomycin‐induced pulmonary fibrosis. Wild‐type mice and mice with high endogenous activated protein C APC levels (APChigh) were subjected to bleomycin‐induced pulmonary fibrosis. Fibrosis was assesses by hydroxyproline and histochemical analysis. Macrophage recruitment was assessed immunohistochemically. In vitro, macrophage migration was analysed by transwell migration assays. Fourteen days after bleomycin instillation, APChigh mice developed pulmonary fibrosis to a similar degree as wild‐type mice. Interestingly, Aschcroft scores as well as lung hydroxyproline levels were significantly lower in APChigh mice than in wild‐type mice on day 28. The reduction in fibrosis in APChigh mice was accompanied by reduced macrophage numbers in their lungs and subsequent in vitro experiments showed that APC inhibits thrombin‐dependent macrophage migration. Our data suggest that high endogenous APC levels inhibit the progression of bleomycin‐induced pulmonary fibrosis and that APC modifies pulmonary fibrosis by limiting thrombin‐dependent macrophage recruitment.
Collapse
Affiliation(s)
- Cong Lin
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Jan von der Thüsen
- Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Berend Isermann
- Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| | - Hartmut Weiler
- Department of Physiology, BloodCenter of Wisconsin, Milwaukee, WI, USA
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Keren Borensztajn
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands.,Inserm UMR1152, Medical School Xavier Bichat, Paris, France.,Département Hospitalo-universtaire FIRE (Fibrosis Inflammation and Remodeling) and LabEx Inflamex, Paris, France
| | - Chris A Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
39
|
Cao YL, Duan Y, Zhu LX, Zhan YN, Min SX, Jin AM. TGF-β1, in association with the increased expression of connective tissue growth factor, induce the hypertrophy of the ligamentum flavum through the p38 MAPK pathway. Int J Mol Med 2016; 38:391-8. [PMID: 27279555 PMCID: PMC4935458 DOI: 10.3892/ijmm.2016.2631] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 05/26/2016] [Indexed: 01/15/2023] Open
Abstract
Hypertrophy of the ligamentum flavum (LF) is one of the key pathomechanisms of lumbar spinal stenosis (LSS). Transforming growth factor (TGF)-β1 is abundantly expressed in hypertrophied degenerative LF tissues from LSS. However, the molecular mechanisms underling the association between TGF-β1 and LF hypertrophy have not yet been fully elucidated. In this study, we investigated the important role of the mitogen-activated protein kinase (MAPK) pathway in the pathogenesis of LSS by analyzing the expression of connective tissue growth factor (CTGF) and extracellular matrix (ECM) components (collagen I and collagen III) in TGF-β1-treated LF cells. Cell growth assay revealed that TGF-β1, in association with CTGF, enhanced the the proliferation of LF cells, and we found that TGF-β1 also elevated CTGF expression and subsequently enhanced the mRNA expression of collagen I and collagen III. The increased mRNA expression levels of CTGF, collagen I and collagen III were abolished by p38 inhibitors. Both immunofluorescence imaging and western blot analysis of p38 and p-p38 revealed the increased expression and phosphorylation of p38. Silencing the expression of p38 by siRNA in LF cells decreased the protein expression of p38, p-p38 and CTGF, as well as the mRNA expression of CTGF, collagen I and collagen III. Taken together, our findings indicate that TGF-β1, in association with the increased expression of CTGF, contribute to the homeostasis of the ECM and to the hypertrophy of LF through the p38 MAPK pathway.
Collapse
Affiliation(s)
- Yan-Lin Cao
- Department of Orthopaedic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Yang Duan
- Department of Orthopaedic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Li-Xin Zhu
- Department of Orthopaedic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Ye-Nan Zhan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Shao-Xiong Min
- Department of Orthopaedic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - An-Min Jin
- Department of Orthopaedic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
40
|
Rosario M, Li F, Wypij D, Roberts AL, Corliss HL, Charlton BM, Frazier AL, Austin SB. Disparities by Sexual Orientation in Frequent Engagement in Cancer-Related Risk Behaviors: A 12-Year Follow-Up. Am J Public Health 2016; 106:698-706. [PMID: 26794176 DOI: 10.2105/ajph.2015.302977] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVES We examined sexual-orientation disparities in frequent engagement in cancer-related risk indicators of tobacco, alcohol, diet and physical activity, ultraviolet radiation, and sexually transmitted infections (STIs). METHODS We used longitudinal data from the national Growing Up Today Study (1999-2010). Of the analytic sample (n = 9958), 1.8% were lesbian or gay (LG), 1.6% bisexual (BI), 12.1% mostly heterosexual (MH), and 84.5% completely heterosexual (CH). RESULTS More sexual minorities (LGs, BIs, and MHs) than CHs frequently engaged in multiple cancer-related risk behaviors (33%, 29%, 28%, and 19%, respectively). Sexual-minority young women, especially BI and MH, more frequently engaged over time in substance use and diet and physical activity risk than CH women. More young gay than CH men frequently engaged over time in vomiting for weight control (odds ratio [OR] = 3.2; 95% confidence interval [CI] = 1.1, 9.4), being physically inactive (OR = 1.7; 95% CI = 1.2, 2.4), and using tanning booths (OR = 4.7; 95% CI = 3.0, 7.4), and had a higher prevalence of ever having an STI (OR = 3.5; 95% CI = 2.0, 6.4). Individual analyses were generally comparable to the group-level analyses. CONCLUSIONS Young sexual minorities are at risk for cancer through frequent exposure to cancer-related risk behaviors over time. Long-term, longitudinal studies and surveillance data are essential and warranted to track frequent engagement in the risk behaviors and cancer-related morbidity and mortality.
Collapse
Affiliation(s)
- Margaret Rosario
- Margaret Rosario is with Department of Psychology, City University of New York-City College and Graduate Center, New York, NY. Fei Li and David Wypij are with Department of Biostatistics, Harvard T. H. Chan School of Public Health (HSPH), Boston, MA. David Wypij, Brittany M. Charlton, A. Lindsay Frazier, and S. Bryn Austin are with Department of Pediatrics, Harvard Medical School (HMS), Boston. David Wypij is also with Department of Cardiology, Boston's Children's Hospital, Boston. Andrea L. Roberts is with Department of Social and Behavioral Sciences, HSPH. Heather L. Corliss is with Division of Health Promotion and Behavioral Science at San Diego State University, San Diego, CA. Brittany M. Charlton and S. Bryn Austin are also with Division of Adolescent and Young Adult Medicine, Boston Children's Hospital. A. Lindsay Frazier is also with Dana-Farber Cancer Institute, Boston, and Department of Epidemiology, HSPH. A. Lindsay Frazier and S. Bryn Austin are also with Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, HMS
| | - Fei Li
- Margaret Rosario is with Department of Psychology, City University of New York-City College and Graduate Center, New York, NY. Fei Li and David Wypij are with Department of Biostatistics, Harvard T. H. Chan School of Public Health (HSPH), Boston, MA. David Wypij, Brittany M. Charlton, A. Lindsay Frazier, and S. Bryn Austin are with Department of Pediatrics, Harvard Medical School (HMS), Boston. David Wypij is also with Department of Cardiology, Boston's Children's Hospital, Boston. Andrea L. Roberts is with Department of Social and Behavioral Sciences, HSPH. Heather L. Corliss is with Division of Health Promotion and Behavioral Science at San Diego State University, San Diego, CA. Brittany M. Charlton and S. Bryn Austin are also with Division of Adolescent and Young Adult Medicine, Boston Children's Hospital. A. Lindsay Frazier is also with Dana-Farber Cancer Institute, Boston, and Department of Epidemiology, HSPH. A. Lindsay Frazier and S. Bryn Austin are also with Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, HMS
| | - David Wypij
- Margaret Rosario is with Department of Psychology, City University of New York-City College and Graduate Center, New York, NY. Fei Li and David Wypij are with Department of Biostatistics, Harvard T. H. Chan School of Public Health (HSPH), Boston, MA. David Wypij, Brittany M. Charlton, A. Lindsay Frazier, and S. Bryn Austin are with Department of Pediatrics, Harvard Medical School (HMS), Boston. David Wypij is also with Department of Cardiology, Boston's Children's Hospital, Boston. Andrea L. Roberts is with Department of Social and Behavioral Sciences, HSPH. Heather L. Corliss is with Division of Health Promotion and Behavioral Science at San Diego State University, San Diego, CA. Brittany M. Charlton and S. Bryn Austin are also with Division of Adolescent and Young Adult Medicine, Boston Children's Hospital. A. Lindsay Frazier is also with Dana-Farber Cancer Institute, Boston, and Department of Epidemiology, HSPH. A. Lindsay Frazier and S. Bryn Austin are also with Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, HMS
| | - Andrea L Roberts
- Margaret Rosario is with Department of Psychology, City University of New York-City College and Graduate Center, New York, NY. Fei Li and David Wypij are with Department of Biostatistics, Harvard T. H. Chan School of Public Health (HSPH), Boston, MA. David Wypij, Brittany M. Charlton, A. Lindsay Frazier, and S. Bryn Austin are with Department of Pediatrics, Harvard Medical School (HMS), Boston. David Wypij is also with Department of Cardiology, Boston's Children's Hospital, Boston. Andrea L. Roberts is with Department of Social and Behavioral Sciences, HSPH. Heather L. Corliss is with Division of Health Promotion and Behavioral Science at San Diego State University, San Diego, CA. Brittany M. Charlton and S. Bryn Austin are also with Division of Adolescent and Young Adult Medicine, Boston Children's Hospital. A. Lindsay Frazier is also with Dana-Farber Cancer Institute, Boston, and Department of Epidemiology, HSPH. A. Lindsay Frazier and S. Bryn Austin are also with Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, HMS
| | - Heather L Corliss
- Margaret Rosario is with Department of Psychology, City University of New York-City College and Graduate Center, New York, NY. Fei Li and David Wypij are with Department of Biostatistics, Harvard T. H. Chan School of Public Health (HSPH), Boston, MA. David Wypij, Brittany M. Charlton, A. Lindsay Frazier, and S. Bryn Austin are with Department of Pediatrics, Harvard Medical School (HMS), Boston. David Wypij is also with Department of Cardiology, Boston's Children's Hospital, Boston. Andrea L. Roberts is with Department of Social and Behavioral Sciences, HSPH. Heather L. Corliss is with Division of Health Promotion and Behavioral Science at San Diego State University, San Diego, CA. Brittany M. Charlton and S. Bryn Austin are also with Division of Adolescent and Young Adult Medicine, Boston Children's Hospital. A. Lindsay Frazier is also with Dana-Farber Cancer Institute, Boston, and Department of Epidemiology, HSPH. A. Lindsay Frazier and S. Bryn Austin are also with Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, HMS
| | - Brittany M Charlton
- Margaret Rosario is with Department of Psychology, City University of New York-City College and Graduate Center, New York, NY. Fei Li and David Wypij are with Department of Biostatistics, Harvard T. H. Chan School of Public Health (HSPH), Boston, MA. David Wypij, Brittany M. Charlton, A. Lindsay Frazier, and S. Bryn Austin are with Department of Pediatrics, Harvard Medical School (HMS), Boston. David Wypij is also with Department of Cardiology, Boston's Children's Hospital, Boston. Andrea L. Roberts is with Department of Social and Behavioral Sciences, HSPH. Heather L. Corliss is with Division of Health Promotion and Behavioral Science at San Diego State University, San Diego, CA. Brittany M. Charlton and S. Bryn Austin are also with Division of Adolescent and Young Adult Medicine, Boston Children's Hospital. A. Lindsay Frazier is also with Dana-Farber Cancer Institute, Boston, and Department of Epidemiology, HSPH. A. Lindsay Frazier and S. Bryn Austin are also with Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, HMS
| | - A Lindsay Frazier
- Margaret Rosario is with Department of Psychology, City University of New York-City College and Graduate Center, New York, NY. Fei Li and David Wypij are with Department of Biostatistics, Harvard T. H. Chan School of Public Health (HSPH), Boston, MA. David Wypij, Brittany M. Charlton, A. Lindsay Frazier, and S. Bryn Austin are with Department of Pediatrics, Harvard Medical School (HMS), Boston. David Wypij is also with Department of Cardiology, Boston's Children's Hospital, Boston. Andrea L. Roberts is with Department of Social and Behavioral Sciences, HSPH. Heather L. Corliss is with Division of Health Promotion and Behavioral Science at San Diego State University, San Diego, CA. Brittany M. Charlton and S. Bryn Austin are also with Division of Adolescent and Young Adult Medicine, Boston Children's Hospital. A. Lindsay Frazier is also with Dana-Farber Cancer Institute, Boston, and Department of Epidemiology, HSPH. A. Lindsay Frazier and S. Bryn Austin are also with Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, HMS
| | - S Bryn Austin
- Margaret Rosario is with Department of Psychology, City University of New York-City College and Graduate Center, New York, NY. Fei Li and David Wypij are with Department of Biostatistics, Harvard T. H. Chan School of Public Health (HSPH), Boston, MA. David Wypij, Brittany M. Charlton, A. Lindsay Frazier, and S. Bryn Austin are with Department of Pediatrics, Harvard Medical School (HMS), Boston. David Wypij is also with Department of Cardiology, Boston's Children's Hospital, Boston. Andrea L. Roberts is with Department of Social and Behavioral Sciences, HSPH. Heather L. Corliss is with Division of Health Promotion and Behavioral Science at San Diego State University, San Diego, CA. Brittany M. Charlton and S. Bryn Austin are also with Division of Adolescent and Young Adult Medicine, Boston Children's Hospital. A. Lindsay Frazier is also with Dana-Farber Cancer Institute, Boston, and Department of Epidemiology, HSPH. A. Lindsay Frazier and S. Bryn Austin are also with Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, HMS
| |
Collapse
|
41
|
Mauro A, Russo V, Di Marcantonio L, Berardinelli P, Martelli A, Muttini A, Mattioli M, Barboni B. M1 and M2 macrophage recruitment during tendon regeneration induced by amniotic epithelial cell allotransplantation in ovine. Res Vet Sci 2016; 105:92-102. [PMID: 27033915 DOI: 10.1016/j.rvsc.2016.01.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 11/25/2015] [Accepted: 01/19/2016] [Indexed: 12/31/2022]
Abstract
Recently, we have demonstrated that ovine amniotic epithelial cells (oAECs) allotransplanted into experimentally induced tendon lesions are able to stimulate tissue regeneration also by reducing leukocyte infiltration. Amongst leukocytes, macrophages (Mφ) M1 and M2 phenotype cells are known to mediate inflammatory and repairing processes, respectively. In this research it was investigated if, during tendon regeneration induced by AECs allotransplantation, M1Mφ and M2Mφ phenotype cells are recruited and differently distributed within the lesion site. Ovine AECs treated and untreated (Ctr) tendons were explanted at 7, 14, and 28 days and tissue microarchitecture was analyzed together with the distribution and quantification of leukocytes (CD45 positive), Mφ (CD68 pan positive), and M1Mφ (CD86, and IL12b) and M2Mφ (CD206, YM1 and IL10) phenotype related markers. In oAEC transplanted tendons CD45 and CD68 positive cells were always reduced in the lesion site. At day 14, oAEC treated tendons began to recover their microarchitecture, contextually a reduction of M1Mφ markers, mainly distributed close to oAECs, and an increase of M2Mφ markers was evidenced. CD206 positive cells were distributed near the regenerating areas. At day 28 oAECs treated tendons acquired a healthy-like structure with a reduction of M2Mφ. Differently, Ctr tendons maintained a disorganized morphology throughout the experimental time and constantly showed high values of M1Mφ markers. These findings indicate that M2Mφ recruitment could be correlated to tendon regeneration induced by oAECs allotransplantation. Moreover, these results demonstrate oAECs immunomodulatory role also in vivo and support novel insights into their allogeneic use underlying the resolution of tendon fibrosis.
Collapse
Affiliation(s)
- Annunziata Mauro
- Faculty of Veterinary Medicine, University of Teramo, Campus Universitario Coste S. Agostino Via R. Balzarini 1, 64100 Teramo, Italy; StemTeCh Group, Italy
| | - Valentina Russo
- Faculty of Veterinary Medicine, University of Teramo, Campus Universitario Coste S. Agostino Via R. Balzarini 1, 64100 Teramo, Italy; StemTeCh Group, Italy.
| | - Lisa Di Marcantonio
- Faculty of Veterinary Medicine, University of Teramo, Campus Universitario Coste S. Agostino Via R. Balzarini 1, 64100 Teramo, Italy
| | - Paolo Berardinelli
- Faculty of Veterinary Medicine, University of Teramo, Campus Universitario Coste S. Agostino Via R. Balzarini 1, 64100 Teramo, Italy
| | - Alessandra Martelli
- Faculty of Veterinary Medicine, University of Teramo, Campus Universitario Coste S. Agostino Via R. Balzarini 1, 64100 Teramo, Italy
| | - Aurelio Muttini
- Faculty of Veterinary Medicine, University of Teramo, Campus Universitario Coste S. Agostino Via R. Balzarini 1, 64100 Teramo, Italy; StemTeCh Group, Italy
| | - Mauro Mattioli
- Faculty of Veterinary Medicine, University of Teramo, Campus Universitario Coste S. Agostino Via R. Balzarini 1, 64100 Teramo, Italy
| | - Barbara Barboni
- Faculty of Veterinary Medicine, University of Teramo, Campus Universitario Coste S. Agostino Via R. Balzarini 1, 64100 Teramo, Italy; StemTeCh Group, Italy
| |
Collapse
|
42
|
Han SY, Choi YJ, Kang MK, Park JHY, Kang YH. Resveratrol Suppresses Cytokine Production Linked to FcεRI-MAPK Activation in IgE-Antigen Complex-Exposed Basophilic Mast Cells and Mice. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 43:1605-23. [DOI: 10.1142/s0192415x15500913] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A complicated interplay between resident mast cells and other recruited inflammatory cells contributes to the development and progression of allergic inflammation entailing the promotion of T helper 2 (Th2) cytokine responses. The current study examined whether resveratrol suppressed the production of inflammatory Th2 cytokines in cultured rat basophilic leukemia RBL-2H3 cells. Cells pre-treated with resveratrol nontoxic at 1–25[Formula: see text][Formula: see text]M were sensitized with anti-dinitrophenyl (anti-DNP), and subsequently stimulated by dinitrophenyl–human serum albumin (DNP–HSA) antigen. Resveratrol dose-dependently diminished the secretion of interleukin (IL)-3, IL-4, IL-13 as well as tumor necrosis factor (TNF)-[Formula: see text] by the antigen stimulation from sensitized cells. It was found that resveratrol mitigated the phosphorylation of p38 MAPK, ERK, and JNK elevated in mast cells exposed to Fc epsilon receptor I (Fc[Formula: see text]RI)-mediated immunoglobulin E (IgE)-antigen complex. The Fc[Formula: see text]RI aggregation was highly enhanced on the surface of mast cells following the HSA stimulation, which was retarded by treatment with 1–25[Formula: see text][Formula: see text]M resveratrol. The IgE-receptor engagement rapidly induced tyrosine phosphorylation of c-Src-related focal adhesion protein paxillin involved in the cytoskeleton rearrangement. The Fc[Formula: see text]RI-mediated rapid activation of c-Src and paxillin was attenuated in a dose-dependent manner. In addition, the paxillin activation entailed p38 MAPK and ERK-responsive signaling, but the JNK activation was less involved. Consistently, oral administration of resveratrol reduced the tissue level of phosphorylated paxillin in the dorsal skin of DNP–HSA-challenged mice. The other tyrosine kinase Tyk2-STAT1 signaling was activated in the dorsal epidermis of antigen-exposed mice, which was associated with allergic inflammation. These results showed that resveratrol inhibited Th2 cytokines- and paxillin-linked allergic responses dependent upon MAPK signaling. Therefore, resveratrol may possess the therapeutic potential of targeting mast cells in preventing the development of allergic inflammation.
Collapse
Affiliation(s)
- Seon-Young Han
- Department of Food Science and Nutrition, Hallym University Chuncheon, Kangwon-do 200-702, Republic of Korea
| | - Yean-Jung Choi
- Department of Food Science and Nutrition, Hallym University Chuncheon, Kangwon-do 200-702, Republic of Korea
| | - Min-Kyung Kang
- Department of Food Science and Nutrition, Hallym University Chuncheon, Kangwon-do 200-702, Republic of Korea
| | - Jung Han Yoon Park
- Department of Food Science and Nutrition, Hallym University Chuncheon, Kangwon-do 200-702, Republic of Korea
| | - Young-Hee Kang
- Department of Food Science and Nutrition, Hallym University Chuncheon, Kangwon-do 200-702, Republic of Korea
| |
Collapse
|
43
|
Jindal A, Bruzzì S, Sutti S, Locatelli I, Bozzola C, Paternostro C, Parola M, Albano E. Fat-laden macrophages modulate lobular inflammation in nonalcoholic steatohepatitis (NASH). Exp Mol Pathol 2015; 99:155-162. [PMID: 26112094 DOI: 10.1016/j.yexmp.2015.06.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/15/2015] [Accepted: 06/19/2015] [Indexed: 12/22/2022]
Abstract
Nonalcoholic steatohepatitis (NASH) is characterized by extensive hepatic monocyte infiltration and monocyte-derived macrophages have an important role in regulating the disease evolution. However, little is known about the functional changes occurring in liver macrophages during NASH progression. In this study, we investigated phenotypic and functional modifications of hepatic macrophages in experimental NASH induced by feeding C57BL/6 mice with a methionine-choline deficient (MCD) diet up to 8weeks. In mice with steatohepatitis liver F4/80-positive macrophages increased in parallel with the disease progression and formed small clusters of enlarged and vacuolated cells. At immunofluorescence these cells contained lipid vesicles positive for the apoptotic cell marker Annexin V suggesting the phagocytosis of apoptotic bodies derived from dead fat-laden hepatocytes. Flow cytometry revealed that these enlarged macrophages expressed inflammatory monocyte (CD11b, Ly6C, TNF-α) markers. However, as compared to regular size macrophages the enlarged sub-set was characterized by an enhanced production of arginase-1 and of the anti-inflammatory mediators IL-10 and annexin A1. Similar vacuolated macrophages producing annexin A1 were also evident in liver biopsies of NASH patients. In mice with NASH, the accumulation of enlarged F4/80(+) cells paralleled with a decline in the expression of the macrophage M1 activation markers iNOS, IL-12 and CXCL10, while the levels of M2 polarization markers arginase-1 and MGL-1 were unchanged. Interestingly, the lowering of IL-12 expression mainly involved the macrophage sub-set with regular size. We conclude that during the progression of NASH fat accumulation within liver macrophages promotes the production of anti-inflammatory mediators that influence hepatic inflammatory responses.
Collapse
Affiliation(s)
- Aastha Jindal
- Dept. of Health Sciences, University "Amedeo Avogadro" of East Piedmont, Novara, Italy; Interdisciplinary Research Centre for Autoimmune Diseases, University "Amedeo Avogadro" of East Piedmont, Novara, Italy
| | - Stefania Bruzzì
- Dept. of Health Sciences, University "Amedeo Avogadro" of East Piedmont, Novara, Italy; Interdisciplinary Research Centre for Autoimmune Diseases, University "Amedeo Avogadro" of East Piedmont, Novara, Italy
| | - Salvatore Sutti
- Dept. of Health Sciences, University "Amedeo Avogadro" of East Piedmont, Novara, Italy; Interdisciplinary Research Centre for Autoimmune Diseases, University "Amedeo Avogadro" of East Piedmont, Novara, Italy
| | - Irene Locatelli
- Dept. of Health Sciences, University "Amedeo Avogadro" of East Piedmont, Novara, Italy; Interdisciplinary Research Centre for Autoimmune Diseases, University "Amedeo Avogadro" of East Piedmont, Novara, Italy
| | - Cristina Bozzola
- Dept. of Health Sciences, University "Amedeo Avogadro" of East Piedmont, Novara, Italy; Interdisciplinary Research Centre for Autoimmune Diseases, University "Amedeo Avogadro" of East Piedmont, Novara, Italy
| | | | - Maurizio Parola
- Dept of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Emanuele Albano
- Dept. of Health Sciences, University "Amedeo Avogadro" of East Piedmont, Novara, Italy; Interdisciplinary Research Centre for Autoimmune Diseases, University "Amedeo Avogadro" of East Piedmont, Novara, Italy.
| |
Collapse
|
44
|
Yamagami K, Oka T, Wang Q, Ishizu T, Lee JK, Miwa K, Akazawa H, Naito AT, Sakata Y, Komuro I. Pirfenidone exhibits cardioprotective effects by regulating myocardial fibrosis and vascular permeability in pressure-overloaded hearts. Am J Physiol Heart Circ Physiol 2015; 309:H512-22. [DOI: 10.1152/ajpheart.00137.2015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/28/2015] [Indexed: 12/12/2022]
Abstract
Although cardiac fibrosis causes heart failure, its molecular mechanisms remain elusive. In this study, we investigated the mechanisms of cardiac fibrosis and examined the effects of the antifibrotic drug pirfenidone (PFD) on chronic heart failure. To understand the responsible mechanisms, we generated an in vivo pressure-overloaded heart failure model via transverse aortic constriction (TAC) and examined the effects of PFD on chronic-phase cardiac fibrosis and function. In the vehicle group, contractile dysfunction and left ventricle fibrosis progressed further from 4 to 8 wk after TAC but were prevented by PFD treatment beginning 4 wk after TAC. We isolated cardiac fibroblasts and vascular endothelial cells from the left ventricles of adult male mice and investigated the cell-type-specific effects of PFD. Transforming growth factor-β induced upregulated collagen 1 expression via p38 phosphorylation and downregulated claudin 5 (Cldn5) expression in cardiac fibroblasts and endothelial cells, respectively; both processes were inhibited by PFD. Moreover, PFD inhibited changes in the collagen 1 and Cldn5 expression levels, resulting in reduced fibrosis and serum albumin leakage into the interstitial space during the chronic phase in TAC hearts. In conclusion, PFD inhibited cardiac fibrosis by suppressing both collagen expression and the increased vascular permeability induced by pressure overload.
Collapse
Affiliation(s)
- Kiyoshi Yamagami
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Toru Oka
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Qi Wang
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Takamaru Ishizu
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Jong-Kook Lee
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Keiko Miwa
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Atsuhiko T. Naito
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- CREST, Japan Science and Technology Agency, Tokyo, Japan
| |
Collapse
|
45
|
Abstract
The unique characteristics of pulmonary circulation and alveolar-epithelial capillary-endothelial barrier allow for maintenance of the air-filled, fluid-free status of the alveoli essential for facilitating gas exchange, maintaining alveolar stability, and defending the lung against inhaled pathogens. The hallmark of pathophysiology in acute respiratory distress syndrome is the loss of the alveolar capillary permeability barrier and the presence of protein-rich edema fluid in the alveoli. This alteration in permeability and accumulation of fluid in the alveoli accompanies damage to the lung epithelium and vascular endothelium along with dysregulated inflammation and inappropriate activity of leukocytes and platelets. In addition, there is uncontrolled activation of coagulation along with suppression of fibrinolysis and loss of surfactant. These pathophysiological changes result in the clinical manifestations of acute respiratory distress syndrome, which include hypoxemia, radiographic opacities, decreased functional residual capacity, increased physiologic deadspace, and decreased lung compliance. Resolution of acute respiratory distress syndrome involves the migration of cells to the site of injury and re-establishment of the epithelium and endothelium with or without the development of fibrosis. Most of the data related to acute respiratory distress syndrome, however, originate from studies in adults or in mature animals with very few studies performed in children or juvenile animals. The lack of studies in children is particularly problematic because the lungs and immune system are still developing during childhood and consequently the pathophysiology of pediatric acute respiratory distress syndrome may differ in significant ways from that seen in acute respiratory distress syndrome in adults. This article describes what is known of the pathophysiologic processes of pediatric acute respiratory distress syndrome as we know it today while also presenting the much greater body of evidence on these processes as elucidated by adult and animal studies. It is also our expressed intent to generate enthusiasm for larger and more in-depth investigations of the mechanisms of disease and repair specific to children in the years to come.
Collapse
|
46
|
Oğuz AK, Yılmaz S, Akar N, Özdağ H, Gürler A, Ateş A, Oygür ÇŞ, Kılıçoğlu SS, Demirtaş S. C-type lectin domain family 12, member A: A common denominator in Behçet's syndrome and acute gouty arthritis. Med Hypotheses 2015; 85:186-91. [PMID: 25957656 DOI: 10.1016/j.mehy.2015.04.032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 04/25/2015] [Indexed: 01/09/2023]
Abstract
C-type lectin domain family 12, member A (CLEC12A) is a C-type lectin-like pattern recognition receptor capable of recognizing monosodium urate crystals. Monosodium urate crystals, the causative agents of gout are also among the danger-associated molecular patterns reflecting cellular injury/cell death. In response to monosodium urate crystals, CLEC12A effectively inhibits granulocyte and monocyte/macrophage functions and hence acts as a negative regulator of inflammation. Behçet's syndrome and gout are autoinflammatory disorders sharing certain pathological (neutrophilic inflammation), clinical (exaggerated response to monosodium urate crystals) and therapeutic (colchicine) features. We propose the hypothesis that decreased expression of CLEC12A is a common denominator in the hyperinflammatory responses observed in Behçet's syndrome and gout. Major lines of evidence supporting this hypothesis are: (1) Downregulation/deficiency of CLEC12A is associated with hyperinflammatory responses. (2) CLEC12A polymorphisms with functional and clinical implications have been documented in other inflammatory diseases. (3) Colchicine, a fundamental therapeutic agent used both in Behçet's syndrome and gout is shown to oppose the downregulation of CLEC12A. (4) Behçet's syndrome and gout are characterized by a hyperinflammatory response to monosodium urate crystals and other than gout, Behçet's syndrome is the only inflammatory condition exhibiting this exaggerated response. (5) Genomewide linkage and association studies of Behçet's syndrome collectively point to 12p12-13, the chromosomal region harboring CLEC12A. (6) Patients with severe forms of Behçet's syndrome underexpress CLEC12A with respect to patients with mild forms of the disease. If supported by well-designed, rigorous experiments, the forementioned hypothesis pertinent to CLEC12A will carry important implications for therapy, designing experimental models, and uncovering immunopathogenic mechanisms in Behçet's syndrome and gout.
Collapse
Affiliation(s)
- Ali Kemal Oğuz
- Department of Internal Medicine, Ufuk University School of Medicine, Ankara, Turkey; Biotechnology Institute, Ankara University, Ankara, Turkey.
| | - Seda Yılmaz
- Department of Internal Medicine, Ufuk University School of Medicine, Ankara, Turkey; Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Nejat Akar
- Department of Internal Medicine, Ufuk University School of Medicine, Ankara, Turkey; Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Hilal Özdağ
- Department of Internal Medicine, Ufuk University School of Medicine, Ankara, Turkey; Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Aysel Gürler
- Department of Internal Medicine, Ufuk University School of Medicine, Ankara, Turkey; Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Aşkın Ateş
- Department of Internal Medicine, Ufuk University School of Medicine, Ankara, Turkey; Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Çağdaş Şahap Oygür
- Department of Internal Medicine, Ufuk University School of Medicine, Ankara, Turkey; Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Sibel Serin Kılıçoğlu
- Department of Internal Medicine, Ufuk University School of Medicine, Ankara, Turkey; Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Selda Demirtaş
- Department of Internal Medicine, Ufuk University School of Medicine, Ankara, Turkey; Biotechnology Institute, Ankara University, Ankara, Turkey
| |
Collapse
|
47
|
Miura K, Egawa Y, Moriki T, Mineta H, Harada H, Baba S, Yamamoto S. Microscopic observation of chemical modification in sections using scanning acoustic microscopy. Pathol Int 2015; 65:355-66. [PMID: 25824722 DOI: 10.1111/pin.12288] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 02/27/2015] [Indexed: 12/01/2022]
Abstract
A scanning acoustic microscope (SAM) calculates the speed of sound (SOS) through tissues and plots the data on the screen to form images. Hard tissues result in greater SOS; based on these differences in tissue properties regarding SOS, SAM can provide data on tissue elasticity. The present study evaluated whether tissue modifications, such as formalin fixation, periodic acid-Schiff (PAS) reactions and protein degradation, changed the acoustic properties of the tissues and whether SAM could be a useful tool for following chemical changes in sections. The fixation process was observable by the increased SOS. During the PAS reaction, the glycosylation of tissues was characterized by an increased SOS. Mucous or glycogen distribution was visualized and was found to be statistically comparable among lesions and states. Protease digestion by pepsin led to a decreased SOS. Tissue sensitivity to proteases varied due to the stage, cause and duration of inflammation or ageing. Changes in acoustic properties were more sensitive than those in optical histology. SAM facilitates the visualisation of the time course or distribution of chemical modifications in tissue sections, thus aiding their comparison among tissues. SAM may be an effective tool for studying changes such as protein cross-linkage, tissue repair and ageing.
Collapse
Affiliation(s)
- Katsutoshi Miura
- Department of Health Science, Pathology and Anatomy, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuki Egawa
- Department of Diagnostic Pathology, Shizuoka City Shizuoka Hospital, Shizuoka, Japan
| | - Toshiaki Moriki
- Department of Diagnostic Pathology, Shizuoka City Shizuoka Hospital, Shizuoka, Japan
| | - Hiroyuki Mineta
- Department of Otorhinolaryngology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hidekazu Harada
- Division of Pathology, Hamamatsu University Hospital, Hamamatsu, Japan
| | - Satoshi Baba
- Division of Pathology, Hamamatsu University Hospital, Hamamatsu, Japan
| | - Seiji Yamamoto
- Medical Photonics Research Center, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
48
|
Keeler GD, Durdik JM, Stenken JA. Localized delivery of dexamethasone-21-phosphate via microdialysis implants in rat induces M(GC) macrophage polarization and alters CCL2 concentrations. Acta Biomater 2015; 12:11-20. [PMID: 25449921 DOI: 10.1016/j.actbio.2014.10.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 09/19/2014] [Accepted: 10/17/2014] [Indexed: 11/27/2022]
Abstract
Microdialysis sampling probes were implanted into the subcutaneous space on the dorsal side of male Sprague Dawley rats to locally deliver dexamethasone-21-phosphate (Dex) with the aim of altering in vivo macrophage polarization. Macrophage polarization is of significant interest in the field of biomaterials since wound-healing macrophages are a possible means to extend implant life as well as improve tissue remodeling to an implant. Quantitative analysis of CCL2 in collected dialysates, gene expression and immunohistochemistry performed on the tissue surrounding the microdialysis implant were used to evaluate if Dex polarized macrophages. Dex infusion down-regulated IL-6 and CCL2 gene expression and decreased CCL2 concentrations in dialysates collected at the implant site. Dex appeared to have no significant effect on the gene regulation of CD163, a commonly used M2c macrophage surface marker; Arg2; and iNOS2. However, Dex infusion was effective at increasing the number of CD163(+) cells surrounding the implanted microdialysis probe. This work demonstrates the use of microdialysis sampling to deliver agents such as Dex to alter macrophage polarization in vivo while allowing the ability to collect cytokines in the surrounding microenvironment.
Collapse
|
49
|
Moriyama K, Kukita A, Li YJ, Uehara N, Zhang JQ, Takahashi I, Kukita T. Regulation of osteoclastogenesis through Tim-3: possible involvement of the Tim-3/galectin-9 system in the modulation of inflammatory bone destruction. J Transl Med 2014; 94:1200-11. [PMID: 25264706 DOI: 10.1038/labinvest.2014.107] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 05/09/2014] [Accepted: 05/19/2014] [Indexed: 11/09/2022] Open
Abstract
Galectins are a unique family of lectins bearing one or two carbohydrate recognition domains (CRDs) that have the ability to bind molecules with β-galactoside-containing carbohydrates. It has been shown that galectins regulate not only cell growth and differentiation but also immune responses, as well as inflammation. Galectin-9, a tandem repeat type of galectin, was originally identified as a chemotactic factor for eosinophils, and is also involved in the regulatory process of inflammation. Here, we examined the involvement of galectin-9 and its receptor, T-cell immunoglobulin- and mucin-domain-containing molecule 3 (Tim-3), in the control of osteoclastogenesis and inflammatory bone destruction. Expression of Tim-3 was detected in osteoclasts and its mononuclear precursors in vivo and in vitro. Galectin-9 markedly inhibited osteoclastogenesis as evaluated in osteoclast precursor cell line RAW-D cells and primary bone marrow cells of mice and rats. The inhibitory effects of galectin-9 on osteoclastogenesis was negated by the addition of β-lactose, an antagonist for galectin binding, suggesting that the inhibitory effect of galectin-9 was mediated through CRD. When galectin-9 was injected into rats with adjuvant-induced arthritis, marked suppression of bone destruction was observed. Inflammatory bone destruction could be efficiently ameliorated by controlling the Tim-3/galectin-9 system in rheumatoid arthritis.
Collapse
Affiliation(s)
- Kanako Moriyama
- 1] Department of Molecular Cell Biology and Oral Anatomy, Faculty of Dental Sciences, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan [2] Department of Orthodontics, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Akiko Kukita
- Department of Microbiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Yin-Ji Li
- Department of Molecular Cell Biology and Oral Anatomy, Faculty of Dental Sciences, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Norihisa Uehara
- Department of Molecular Cell Biology and Oral Anatomy, Faculty of Dental Sciences, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Jing-Qi Zhang
- Department of Molecular Cell Biology and Oral Anatomy, Faculty of Dental Sciences, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ichiro Takahashi
- Department of Orthodontics, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology and Oral Anatomy, Faculty of Dental Sciences, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
50
|
Keightley MC, Wang CH, Pazhakh V, Lieschke GJ. Delineating the roles of neutrophils and macrophages in zebrafish regeneration models. Int J Biochem Cell Biol 2014; 56:92-106. [DOI: 10.1016/j.biocel.2014.07.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/18/2014] [Accepted: 07/14/2014] [Indexed: 12/24/2022]
|