251
|
Marion T, Elbahesh H, Thomas PG, DeVincenzo JP, Webby R, Schughart K. Respiratory Mucosal Proteome Quantification in Human Influenza Infections. PLoS One 2016; 11:e0153674. [PMID: 27088501 PMCID: PMC4835085 DOI: 10.1371/journal.pone.0153674] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/01/2016] [Indexed: 01/08/2023] Open
Abstract
Respiratory influenza virus infections represent a serious threat to human health. Underlying medical conditions and genetic make-up predispose some influenza patients to more severe forms of disease. To date, only a few studies have been performed in patients to correlate a selected group of cytokines and chemokines with influenza infection. Therefore, we evaluated the potential of a novel multiplex micro-proteomics technology, SOMAscan, to quantify proteins in the respiratory mucosa of influenza A and B infected individuals. The analysis included but was not limited to quantification of cytokines and chemokines detected in previous studies. SOMAscan quantified more than 1,000 secreted proteins in small nasal wash volumes from infected and healthy individuals. Our results illustrate the utility of micro-proteomic technology for analysis of proteins in small volumes of respiratory mucosal samples. Furthermore, when we compared nasal wash samples from influenza-infected patients with viral load ≥ 28 and increased IL-6 and CXCL10 to healthy controls, we identified 162 differentially-expressed proteins between the two groups. This number greatly exceeds the number of DEPs identified in previous studies in human influenza patients. Most of the identified proteins were associated with the host immune response to infection, and changes in protein levels of 151 of the DEPs were significantly correlated with viral load. Most important, SOMAscan identified differentially expressed proteins heretofore not associated with respiratory influenza infection in humans. Our study is the first report for the use of SOMAscan to screen nasal secretions. It establishes a precedent for micro-proteomic quantification of proteins that reflect ongoing response to respiratory infection.
Collapse
Affiliation(s)
- Tony Marion
- University of Tennessee Health Science Center, Department of Microbiology, Immunology and Biochemistry, Memphis, United States of America
| | - Husni Elbahesh
- University of Tennessee Health Science Center, Department of Microbiology, Immunology and Biochemistry, Memphis, United States of America
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, United States of America
| | - John P. DeVincenzo
- University of Tennessee Health Science Center, Department of Microbiology, Immunology and Biochemistry, Memphis, United States of America
- University of Tennessee Health Science Center, Department of Pediatrics, Memphis, United States of America
- Children’s Foundation Research Center at Le Bonheur Children’s Hospital, Memphis, United States of America
| | - Richard Webby
- Department of Infectious Diseases, St Jude Children’s Research Hospital, Memphis, United States of America
| | - Klaus Schughart
- University of Tennessee Health Science Center, Department of Microbiology, Immunology and Biochemistry, Memphis, United States of America
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
- University of Veterinary Medicine Hannover, Hannover, Germany
- * E-mail:
| |
Collapse
|
252
|
Machado ID, Spatti M, Hastreiter A, Santin JR, Fock RA, Gil CD, Oliani SM, Perretti M, Farsky SHP. Annexin A1 Is a Physiological Modulator of Neutrophil Maturation and Recirculation Acting on the CXCR4/CXCL12 Pathway. J Cell Physiol 2016; 231:2418-27. [PMID: 26892496 DOI: 10.1002/jcp.25346] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/16/2016] [Indexed: 12/18/2022]
Abstract
Neutrophil production and traffic in the body compartments is finely controlled, and the strong evidences support the role of CXCL12/CXCR4 pathway on neutrophil trafficking to and from the bone marrow (BM). We recently showed that the glucocorticoid-regulated protein, Annexin A1 (AnxA1) modulates neutrophil homeostasis and here we address the effects of AnxA1 on steady-state neutrophil maturation and trafficking. For this purpose, AnxA1(-/-) and Balb/C wild-type mice (WT) were donors of BM granulocytes and mesenchymal stem cells and blood neutrophils. In vivo treatments with the pharmacological AnxA1 mimetic peptide (Ac2-26) or the formyl peptide receptor (FPR) antagonist (Boc-2) were used to elucidate the pathway of AnxA1 action, and with the cytosolic glucocorticoid antagonist receptor RU 38486. Accelerated maturation of BM granulocytes was detected in AnxA1(-/-) and Boc2-treated WT mice, and was reversed by treatment with Ac2-26 in AnxA1(-/-) mice. AnxA1 and FPR2 were constitutively expressed in bone marrow granulocytes, and their expressions were reduced by treatment with RU38486. Higher numbers of CXCR4(+) neutrophils were detected in the circulation of AnxA1(-/-) or Boc2-treated WT mice, and values were rescued in Ac2-26-treated AnxA1(-/-) mice. Although circulating neutrophils of AnxA1(-/-) animals were CXCR4(+) , they presented reduced CXCL12-induced chemotaxis. Moreover, levels of CXCL12 were reduced in the bone marrow perfusate and in the mesenchymal stem cell supernatant from AnxA1(-/-) mice, and in vivo and in vitro CXCL12 expression was re-established after Ac2-26 treatment. Collectively, these data highlight AnxA1 as a novel determinant of neutrophil maturation and the mechanisms behind blood neutrophil homing to BM via the CXCL12/CXCR4 pathway. J. Cell. Physiol. 231: 2418-2427, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Isabel Daufenback Machado
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Marina Spatti
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Araceli Hastreiter
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - José Roberto Santin
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Ricardo Ambrósio Fock
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Cristiane Damas Gil
- Department of Morphology and Genetics, Federal University of São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Sonia Maria Oliani
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), São Paulo State University (UNESP), São José do Rio Preto, São Paulo, Brazil
| | - Mauro Perretti
- Centre for Biochemical Pharmacology, The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, London, United Kingdom
| | - Sandra Helena Poliselli Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
253
|
Grabiec AM, Hussell T. The role of airway macrophages in apoptotic cell clearance following acute and chronic lung inflammation. Semin Immunopathol 2016; 38:409-23. [PMID: 26957481 PMCID: PMC4896990 DOI: 10.1007/s00281-016-0555-3] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/29/2016] [Indexed: 12/19/2022]
Abstract
Acute and chronic inflammatory responses in the lung are associated with the accumulation of large quantities of immune and structural cells undergoing apoptosis, which need to be engulfed by phagocytes in a process called ‘efferocytosis’. Apoptotic cell recognition and removal from the lung is mediated predominantly by airway macrophages, though immature dendritic cells and non-professional phagocytes, such as epithelial cells and mesenchymal cells, can also display this function. Efficient clearance of apoptotic cells from the airways is essential for successful resolution of inflammation and the return to lung homeostasis. Disruption of this process leads to secondary necrosis of accumulating apoptotic cells, release of necrotic cell debris and subsequent uncontrolled inflammatory activation of the innate immune system by the released ‘damage associated molecular patterns’ (DAMPS). To control the duration of the immune response and prevent autoimmune reactions, anti-inflammatory signalling cascades are initiated in the phagocyte upon apoptotic cell uptake, mediated by a range of receptors that recognise specific phospholipids or proteins externalised on, or secreted by, the apoptotic cell. However, prolonged activation of apoptotic cell recognition receptors, such as the family of receptor tyrosine kinases Tyro3, Axl and MerTK (TAM), may delay or prevent inflammatory responses to subsequent infections. In this review, we will discuss recent advances in our understanding of the mechanism controlling apoptotic cell recognition and removal from the lung in homeostasis and during inflammation, the contribution of defective efferocytosis to chronic inflammatory lung diseases, such as chronic obstructive pulmonary disease, asthma and cystic fibrosis, and implications of the signals triggered by apoptotic cells in the susceptibility to pulmonary microbial infections.
Collapse
Affiliation(s)
- Aleksander M Grabiec
- Manchester Collaborative Centre for Inflammation Research, Core Technology Facility, The University of Manchester, 46 Grafton Street, M13 9NT, Manchester, UK
| | - Tracy Hussell
- Manchester Collaborative Centre for Inflammation Research, Core Technology Facility, The University of Manchester, 46 Grafton Street, M13 9NT, Manchester, UK.
| |
Collapse
|
254
|
Martindale RG, Warren MM, McClave SA. Does the use of specialized proresolving molecules in critical care offer a more focused approach to controlling inflammation than that of fish oils? Curr Opin Clin Nutr Metab Care 2016; 19:151-4. [PMID: 26828585 DOI: 10.1097/mco.0000000000000250] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW The literature regarding the use of fish oils in the critically ill to limit the inflammatory and catabolic response have been inconsistent. The objective of this manuscript is to review a newly discovered class of specialized proresolving molecules (SPMs), which could help elucidate the discrepancies reported in the critical care literature regarding the anti-inflammatory benefits of fish oil/ω-3 fatty acids. RECENT FINDINGS Although use of fish oil has traditionally been thought to reduce or limit the inflammatory process in the critical ill, a new class of endogenously produced highly active lipid mediators derived from arachidonic acid and ω-3 fatty acids (lipoxins, resolvins, protectins, and maresins) have been shown to actively enhance resolution of inflammation. These SPMs stimulate the cardinal signs of resolution of inflammation, which include the cessation of leukocytic infiltration, a countering of the effects of proinflammatory mediators, stimulation of the uptake of apoptotic neutrophils, promotion of the clearance of necrotic cellular debris, and enhancement of the host's ability to eliminate microbial invasion. SUMMARY By actively turning off inflammation (instead of simply attenuating its natural course), SPMs have shown more consistent effects in decreasing pain and risk of sepsis, increasing epithelialization and wound healing, promoting tissue regeneration, potentiating the effects of antibiotics, and enhancing adaptive immunity.
Collapse
Affiliation(s)
- Robert G Martindale
- aDepartment of Surgery, Oregon Health and Science University, Portland, Oregon bDepartment of Medicine, University of Louisville, Louisville, Kentucky, USA
| | | | | |
Collapse
|
255
|
Biomaterials approaches to modeling macrophage-extracellular matrix interactions in the tumor microenvironment. Curr Opin Biotechnol 2016; 40:16-23. [PMID: 26921768 DOI: 10.1016/j.copbio.2016.02.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 01/12/2023]
Abstract
Tumors are characterized by aberrant extracellular matrix (ECM) remodeling and chronic inflammation. While advances in biomaterials and tissue engineering strategies have led to important new insights regarding the role of ECM composition, structure, and mechanical properties in cancer in general, the functional link between these parameters and macrophage phenotype is poorly understood. Nevertheless, increasing experimental evidence suggests that macrophage behavior is similarly controlled by physicochemical properties of the ECM and consequential changes in mechanosignaling. Here, we will summarize the current knowledge of macrophage biology and ECM-mediated differences in mechanotransduction and discuss future opportunities of biomaterials and tissue engineering platforms to interrogate the functional relationship between these parameters and their relevance to cancer.
Collapse
|
256
|
Gu Z, Lamont GJ, Lamont RJ, Uriarte SM, Wang H, Scott DA. Resolvin D1, resolvin D2 and maresin 1 activate the GSK3β anti-inflammatory axis in TLR4-engaged human monocytes. Innate Immun 2016; 22:186-95. [PMID: 26878867 DOI: 10.1177/1753425916628618] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/31/2015] [Indexed: 01/08/2023] Open
Abstract
Pro-resolving, docosahexaenoic acid-derived mediators have recently emerged as important potential therapeutic agents for the amelioration of complications arising from inflammation, such as vascular disease, asthma, acute lung injury and colitis. While resolvin D1 (RVD1), resolvin D2 (RVD2) and maresin 1 (MaR1) are established pro-resolvins, their mechanisms of action remain unclear. Here we show that, in LPS-stimulated primary human monocytes, RVD1, RVD2 and MaR1 each suppress the release of pro-inflammatory cytokines (TNF, IL-1β, IL-8) and the innate/adaptive bridging cytokine, IL-12 p40, while simultaneously augmenting the production of the anti-inflammatory cytokine, IL-10. Such resolving activity is accompanied by the increased phosphorylation (enhanced anti-inflammatory state) of glycogen synthase kinase 3β (GSK3β) along with increased phosphorylation (activation) of Akt, SGK1 and CREB but not MAPK-related molecules. Gain and loss of function experiments confirm a key role for GSK3β and CREB in the anti-inflammatory actions of resolvins. These results suggest that induction of the GSK3β anti-inflammatory axis is a common mechanism of action for RVD1, RVD2 and MaR1.
Collapse
Affiliation(s)
- Zhen Gu
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Gwyneth J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - Silvia M Uriarte
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA Department of Medicine, University of Louisville, KY, USA
| | - Huizhi Wang
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| | - David A Scott
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, KY, USA
| |
Collapse
|
257
|
Padera TP, Meijer EFJ, Munn LL. The Lymphatic System in Disease Processes and Cancer Progression. Annu Rev Biomed Eng 2016; 18:125-58. [PMID: 26863922 DOI: 10.1146/annurev-bioeng-112315-031200] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Advances in our understanding of the structure and function of the lymphatic system have made it possible to identify its role in a variety of disease processes. Because it is involved not only in fluid homeostasis but also in immune cell trafficking, the lymphatic system can mediate and ultimately alter immune responses. Our rapidly increasing knowledge of the molecular control of the lymphatic system will inevitably lead to new and effective therapies for patients with lymphatic dysfunction. In this review, we discuss the molecular and physiological control of lymphatic vessel function and explore how the lymphatic system contributes to many disease processes, including cancer and lymphedema.
Collapse
Affiliation(s)
- Timothy P Padera
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| | - Eelco F J Meijer
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| |
Collapse
|
258
|
Prockop DJ. Inflammation, fibrosis, and modulation of the process by mesenchymal stem/stromal cells. Matrix Biol 2016; 51:7-13. [PMID: 26807758 DOI: 10.1016/j.matbio.2016.01.010] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Fibrosis and scarring are the end stage of many disease processes. In effect, the collagen fibers that initially provide a necessary strength during the repair of injured tissues are frequently synthesized in excessive amounts and become irreversible fibrotic deposits that limit regeneration of the endogenous cells of a tissue. This review will focus on the potential of mesenchymal stem/stromal cells for treatment of fibrotic diseases, with emphasis on the role of TSG-6 as a mediator of anti-inflammatory effects.
Collapse
Affiliation(s)
- Darwin J Prockop
- Institute for Regenerative Medicine, Texas A&M University, College of Medicine, Temple, TX, USA.
| |
Collapse
|
259
|
Annexin A1 and the Resolution of Inflammation: Modulation of Neutrophil Recruitment, Apoptosis, and Clearance. J Immunol Res 2016; 2016:8239258. [PMID: 26885535 PMCID: PMC4738713 DOI: 10.1155/2016/8239258] [Citation(s) in RCA: 222] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 12/01/2015] [Indexed: 12/13/2022] Open
Abstract
Neutrophils (also named polymorphonuclear leukocytes or PMN) are essential components of the immune system, rapidly recruited to sites of inflammation, providing the first line of defense against invading pathogens. Since neutrophils can also cause tissue damage, their fine-tuned regulation at the inflammatory site is required for proper resolution of inflammation. Annexin A1 (AnxA1), also known as lipocortin-1, is an endogenous glucocorticoid-regulated protein, which is able to counterregulate the inflammatory events restoring homeostasis. AnxA1 and its mimetic peptides inhibit neutrophil tissue accumulation by reducing leukocyte infiltration and activating neutrophil apoptosis. AnxA1 also promotes monocyte recruitment and clearance of apoptotic leukocytes by macrophages. More recently, some evidence has suggested the ability of AnxA1 to induce macrophage reprogramming toward a resolving phenotype, resulting in reduced production of proinflammatory cytokines and increased release of immunosuppressive and proresolving molecules. The combination of these mechanisms results in an effective resolution of inflammation, pointing to AnxA1 as a promising tool for the development of new therapeutic strategies to treat inflammatory diseases.
Collapse
|
260
|
Balas L, Durand T. Dihydroxylated E,E,Z-docosatrienes. An overview of their synthesis and biological significance. Prog Lipid Res 2016; 61:1-18. [DOI: 10.1016/j.plipres.2015.10.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 10/21/2015] [Accepted: 10/28/2015] [Indexed: 12/20/2022]
|
261
|
Möller K, Pösel C, Kranz A, Schulz I, Scheibe J, Didwischus N, Boltze J, Weise G, Wagner DC. Arterial Hypertension Aggravates Innate Immune Responses after Experimental Stroke. Front Cell Neurosci 2015; 9:461. [PMID: 26640428 PMCID: PMC4661280 DOI: 10.3389/fncel.2015.00461] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/12/2015] [Indexed: 01/13/2023] Open
Abstract
Arterial hypertension is not only the leading risk factor for stroke, but also attributes to impaired recovery and poor outcome. The latter could be explained by hypertensive vascular remodeling that aggravates perfusion deficits and blood–brain barrier disruption. However, besides vascular changes, one could hypothesize that activation of the immune system due to pre-existing hypertension may negatively influence post-stroke inflammation and thus stroke outcome. To test this hypothesis, male adult spontaneously hypertensive rats (SHRs) and normotensive Wistar Kyoto rats (WKYs) were subjected to photothrombotic stroke. One and 3 days after stroke, infarct volume and functional deficits were evaluated by magnetic resonance imaging and behavioral tests. Expression levels of adhesion molecules and chemokines along with the post-stroke inflammatory response were analyzed by flow cytometry, quantitative real-time PCR and immunohistochemistry in rat brains 4 days after stroke. Although comparable at day 1, lesion volumes were significantly larger in SHR at day 3. The infarct volume showed a strong correlation with the amount of CD45 highly positive leukocytes present in the ischemic hemispheres. Functional deficits were comparable between SHR and WKY. Brain endothelial expression of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), and P-selectin (CD62P) was neither increased by hypertension nor by stroke. However, in SHR, brain infiltrating myeloid leukocytes showed significantly higher surface expression of ICAM-1 which may augment leukocyte transmigration by leukocyte–leukocyte interactions. The expression of chemokines that primarily attract monocytes and granulocytes was significantly increased by stroke and, furthermore, by hypertension. Accordingly, ischemic hemispheres of SHR contain considerably higher numbers of monocytes, macrophages and granulocytes. Exacerbated brain inflammation in SHR may finally be responsible for larger infarct volumes. These findings provide an immunological explanation for the epidemiological observation that existing hypertension negatively affects stroke outcome and mortality.
Collapse
Affiliation(s)
- Karoline Möller
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany ; Institute of Anatomy, Histology and Embryology, University of Leipzig Leipzig, Germany
| | - Claudia Pösel
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany
| | - Alexander Kranz
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany
| | - Isabell Schulz
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany
| | - Johanna Scheibe
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany
| | - Nadine Didwischus
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany ; Research Group Human Biology, Institute of Biology, University of Leipzig Leipzig, Germany
| | - Johannes Boltze
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany ; Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck Lübeck, Germany
| | - Gesa Weise
- Fraunhofer Institute for Cell Therapy and Immunology Leipzig, Germany ; Department of Neurology, University of Leipzig Leipzig, Germany
| | | |
Collapse
|
262
|
Headland SE, Jones HR, Norling LV, Kim A, Souza PR, Corsiero E, Gil CD, Nerviani A, Dell'Accio F, Pitzalis C, Oliani SM, Jan LY, Perretti M. Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis. Sci Transl Med 2015; 7:315ra190. [PMID: 26606969 PMCID: PMC6034622 DOI: 10.1126/scitranslmed.aac5608] [Citation(s) in RCA: 230] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Microvesicles (MVs) are emerging as a new mechanism of intercellular communication by transferring cellular lipid and protein components to target cells, yet their function in disease is only now being explored. We found that neutrophil-derived MVs were increased in concentration in synovial fluid from rheumatoid arthritis patients compared to paired plasma. Synovial MVs overexpressed the proresolving, anti-inflammatory protein annexin A1 (AnxA1). Mice deficient in TMEM16F, a lipid scramblase required for microvesiculation, exhibited exacerbated cartilage damage when subjected to inflammatory arthritis. To determine the function of MVs in inflammatory arthritis, toward the possibility of MV-based therapeutics, we examined the role of immune cell-derived MVs in rodent models and in human primary chondrocytes. In vitro, exogenous neutrophil-derived AnxA1(+) MVs activated anabolic gene expression in chondrocytes, leading to extracellular matrix accumulation and cartilage protection through the reduction in stress-adaptive homeostatic mediators interleukin-8 and prostaglandin E2. In vivo, intra-articular injection of AnxA1(+) MV lessened cartilage degradation caused by inflammatory arthritis. Arthritic mice receiving adoptive transfer of whole neutrophils displayed abundant MVs within cartilage matrix and revealed that MVs, but not neutrophils themselves, can penetrate cartilage. Mechanistic studies support a model whereby MV-associated AnxA1 interacts with its receptor FPR2 (formyl peptide receptor 2)/ALX, increasing transforming growth factor-β production by chondrocytes, ultimately leading to cartilage protection. We envisage that MVs, either directly or loaded with therapeutics, can be harnessed as a unique therapeutic strategy for protection in diseases associated with cartilage degeneration.
Collapse
Affiliation(s)
- Sarah E Headland
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Hefin R Jones
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Lucy V Norling
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Andrew Kim
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Patricia R Souza
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Elisa Corsiero
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Cristiane D Gil
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas, São Paulo State University (UNESP), São José do Rio Preto 15054-000, Brazil
| | - Alessandra Nerviani
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Francesco Dell'Accio
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Department of Rheumatology, Barts Health Trust, Bancroft Road, London E1 4DG, UK
| | - Costantino Pitzalis
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK. Department of Rheumatology, Barts Health Trust, Bancroft Road, London E1 4DG, UK
| | - Sonia M Oliani
- Department of Biology, Instituto de Biociências, Letras e Ciências Exatas, São Paulo State University (UNESP), São José do Rio Preto 15054-000, Brazil
| | - Lily Y Jan
- Department of Physiology, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| |
Collapse
|
263
|
He ZX, Chen XW, Zhou ZW, Zhou SF. Impact of physiological, pathological and environmental factors on the expression and activity of human cytochrome P450 2D6 and implications in precision medicine. Drug Metab Rev 2015; 47:470-519. [PMID: 26574146 DOI: 10.3109/03602532.2015.1101131] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
With only 1.3-4.3% in total hepatic CYP content, human CYP2D6 can metabolize more than 160 drugs. It is a highly polymorphic enzyme and subject to marked inhibition by a number of drugs, causing a large interindividual variability in drug clearance and drug response and drug-drug interactions. The expression and activity of CYP2D6 are regulated by a number of physiological, pathological and environmental factors at transcriptional, post-transcriptional, translational and epigenetic levels. DNA hypermethylation and histone modifications can repress the expression of CYP2D6. Hepatocyte nuclear factor-4α binds to a directly repeated element in the promoter of CYP2D6 and thus regulates the expression of CYP2D6. Small heterodimer partner represses hepatocyte nuclear factor-4α-mediated transactivation of CYP2D6. GW4064, a farnesoid X receptor agonist, decreases hepatic CYP2D6 expression and activity while increasing small heterodimer partner expression and its recruitment to the CYP2D6 promoter. The genotypes are key determinants of interindividual variability in CYP2D6 expression and activity. Recent genome-wide association studies have identified a large number of genes that can regulate CYP2D6. Pregnancy induces CYP2D6 via unknown mechanisms. Renal or liver diseases, smoking and alcohol use have minor to moderate effects only on CYP2D6 activity. Unlike CYP1 and 3 and other CYP2 members, CYP2D6 is resistant to typical inducers such as rifampin, phenobarbital and dexamethasone. Post-translational modifications such as phosphorylation of CYP2D6 Ser135 have been observed, but the functional impact is unknown. Further functional and validation studies are needed to clarify the role of nuclear receptors, epigenetic factors and other factors in the regulation of CYP2D6.
Collapse
Affiliation(s)
- Zhi-Xu He
- a Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center & Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University , Guiyang , Guizhou , China
| | - Xiao-Wu Chen
- b Department of General Surgery , The First People's Hospital of Shunde, Southern Medical University , Shunde , Foshan , Guangdong , China , and
| | - Zhi-Wei Zhou
- c Department of Pharmaceutical Science , College of Pharmacy, University of South Florida , Tampa , FL , USA
| | - Shu-Feng Zhou
- a Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center & Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University , Guiyang , Guizhou , China .,c Department of Pharmaceutical Science , College of Pharmacy, University of South Florida , Tampa , FL , USA
| |
Collapse
|
264
|
Assessment of β-lapachone loaded in lecithin-chitosan nanoparticles for the topical treatment of cutaneous leishmaniasis in L. major infected BALB/c mice. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:2003-12. [DOI: 10.1016/j.nano.2015.07.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 06/22/2015] [Accepted: 07/17/2015] [Indexed: 12/23/2022]
|
265
|
Uthayashanker RE, Rita MH. Preliminary screening of anti-inflammatory effect of phytochemicals on chemotaxis of human neutrophils. ACTA ACUST UNITED AC 2015. [DOI: 10.5897/jpp2015.0353] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
266
|
Gilroy D, De Maeyer R. New insights into the resolution of inflammation. Semin Immunol 2015; 27:161-8. [PMID: 26037968 DOI: 10.1016/j.smim.2015.05.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/06/2015] [Accepted: 05/13/2015] [Indexed: 12/31/2022]
Abstract
The goal of treating chronic inflammatory diseases must be to inhibit persistent inflammation and restore tissue function. To achieve this we need to improve our understanding of the pathways that drive inflammation as well as those that bring about its resolution. In particular, resolution of inflammation is driven by a complex set of mediators that regulate cellular events required to clear inflammatory cells from sites of injury or infection and restore homeostasis. Indeed, it may be argued that dysfunctional resolution may underpin the aetiology of some chronic inflammatory disease and that a novel goal in treating such diseases is to develop drugs based on the mode of endogenous pro-resolution factors in order to drive on-going inflammation down a pro-resolution pathway. And while we are improving our understanding of the resolution of acute and chronic inflammation, much remains to be discovered. Here we will discuss the key endogenous checkpoints necessary for mounting an effective yet limited inflammatory response and the crucial biochemical pathways necessary to prevent its persistence and trigger its resolution. In doing so, we will provide an update on what is known about resolution of acute inflammation, in particular its link with adaptive immunity.
Collapse
Affiliation(s)
- Derek Gilroy
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street, University College London, London WC1E 6JJ, United Kingdom.
| | - Roel De Maeyer
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street, University College London, London WC1E 6JJ, United Kingdom
| |
Collapse
|