601
|
Thieblemont N, Wright HL, Edwards SW, Witko-Sarsat V. Human neutrophils in auto-immunity. Semin Immunol 2016; 28:159-73. [DOI: 10.1016/j.smim.2016.03.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 03/08/2016] [Accepted: 03/12/2016] [Indexed: 01/06/2023]
|
602
|
Van Avondt K, van der Linden M, Naccache PH, Egan DA, Meyaard L. Signal Inhibitory Receptor on Leukocytes-1 Limits the Formation of Neutrophil Extracellular Traps, but Preserves Intracellular Bacterial Killing. THE JOURNAL OF IMMUNOLOGY 2016; 196:3686-94. [DOI: 10.4049/jimmunol.1501650] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 03/03/2016] [Indexed: 11/19/2022]
|
603
|
McGuinness WA, Kobayashi SD, DeLeo FR. Evasion of Neutrophil Killing by Staphylococcus aureus. Pathogens 2016; 5:E32. [PMID: 26999220 PMCID: PMC4810153 DOI: 10.3390/pathogens5010032] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/11/2016] [Accepted: 03/14/2016] [Indexed: 01/08/2023] Open
Abstract
Staphylococcus aureus causes many types of infections, ranging from self-resolving skin infections to severe or fatal pneumonia. Human innate immune cells, called polymorphonuclear leukocytes (PMNs or neutrophils), are essential for defense against S. aureus infections. Neutrophils are the most prominent cell type of the innate immune system and are capable of producing non-specific antimicrobial molecules that are effective at eliminating bacteria. Although significant progress has been made over the past few decades, our knowledge of S. aureus-host innate immune system interactions is incomplete. Most notably, S. aureus has the capacity to produce numerous molecules that are directed to protect the bacterium from neutrophils. Here we review in brief the role played by neutrophils in defense against S. aureus infection, and correspondingly, highlight selected S. aureus molecules that target key neutrophil functions.
Collapse
Affiliation(s)
- Will A McGuinness
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA.
| | - Scott D Kobayashi
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA.
| | - Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 903 South 4th Street, Hamilton, MT 59840, USA.
| |
Collapse
|
604
|
van Rees DJ, Szilagyi K, Kuijpers TW, Matlung HL, van den Berg TK. Immunoreceptors on neutrophils. Semin Immunol 2016; 28:94-108. [PMID: 26976825 PMCID: PMC7129252 DOI: 10.1016/j.smim.2016.02.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 02/24/2016] [Accepted: 02/26/2016] [Indexed: 12/12/2022]
Abstract
Neutrophil activities must be tightly controlled to maintain immune homeostasis. Activating and inhibitory receptors balance the outcome of immune cell activation. Immunoreceptors contain Ig-like extracellular domains and signal via ITAMs or ITIMs. Syk or SHP/SHIP mediate downstream signaling after immunoreceptor activation. Targeting immunoreceptors provides opportunities for therapeutic interventions.
Neutrophils play a critical role in the host defense against infection, and they are able to perform a variety of effector mechanisms for this purpose. However, there are also a number of pathological conditions, including autoimmunity and cancer, in which the activities of neutrophils can be harmful to the host. Thus the activities of neutrophils need to be tightly controlled. As in the case of other immune cells, many of the neutrophil effector functions are regulated by a series of immunoreceptors on the plasma membrane. Here, we review what is currently known about the functions of the various individual immunoreceptors and their signaling in neutrophils. While these immunoreceptors allow for the recognition of a diverse range of extracellular ligands, such as cell surface structures (like proteins, glycans and lipids) and extracellular matrix components, they commonly signal via conserved ITAM or ITIM motifs and their associated downstream pathways that depend on the phosphorylation of tyrosine residues in proteins and/or inositol lipids. This allows for a balanced homeostatic regulation of neutrophil effector functions. Given the number of available immunoreceptors and their fundamental importance for neutrophil behavior, it is perhaps not surprising that pathogens have evolved means to evade immune responses through some of these pathways. Inversely, some of these receptors evolved to specifically recognize these pathogens. Finally, some interactions mediated by immunoreceptors in neutrophils have been identified as promising targets for therapeutic intervention.
Collapse
Affiliation(s)
- Dieke J van Rees
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Katka Szilagyi
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timo K van den Berg
- Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
605
|
Jhunjhunwala S, Alvarez D, Aresta-DaSilva S, Tang K, Tang BC, Greiner DL, Newburger PE, von Andrian UH, Langer R, Anderson DG. Frontline Science: Splenic progenitors aid in maintaining high neutrophil numbers at sites of sterile chronic inflammation. J Leukoc Biol 2016; 100:253-60. [PMID: 26965635 DOI: 10.1189/jlb.1hi0615-248rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 02/04/2016] [Indexed: 12/30/2022] Open
Abstract
Neutrophils are constantly generated from hematopoietic stem and progenitor cells in the bone marrow to maintain high numbers in circulation. A considerable number of neutrophils and their progenitors have been shown to be present in the spleen too; however, their exact role in this organ remains unclear. Herein, we sought to study the function of splenic neutrophils and their progenitors using a mouse model for sterile, peritoneal inflammation. In this microcapsule device implantation model, we show chronic neutrophil presence at implant sites, with recruitment from circulation as the primary mechanism for their prevalence in the peritoneal exudate. Furthermore, we demonstrate that progenitor populations in the spleen play a key role in maintaining elevated neutrophil numbers. Our results provide new insight into the role for splenic neutrophils and their progenitors and establish a model to study neutrophil function during sterile inflammation.
Collapse
Affiliation(s)
- Siddharth Jhunjhunwala
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - David Alvarez
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Stephanie Aresta-DaSilva
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Katherine Tang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Benjamin C Tang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Dale L Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Peter E Newburger
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA; Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA; and
| | - Ulrich H von Andrian
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA; The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge, Massachusetts, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Harvard-Massachusetts Institute of Technology Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Anesthesiology, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Harvard-Massachusetts Institute of Technology Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA;
| |
Collapse
|
606
|
Carevic M, Öz H, Fuchs K, Laval J, Schroth C, Frey N, Hector A, Bilich T, Haug M, Schmidt A, Autenrieth SE, Bucher K, Beer-Hammer S, Gaggar A, Kneilling M, Benarafa C, Gao JL, Murphy PM, Schwarz S, Moepps B, Hartl D. CXCR1 Regulates Pulmonary Anti-Pseudomonas Host Defense. J Innate Immun 2016; 8:362-73. [PMID: 26950764 DOI: 10.1159/000444125] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/19/2016] [Indexed: 11/19/2022] Open
Abstract
Pseudomonas aeruginosa is a key opportunistic pathogen causing disease in cystic fibrosis (CF) and other lung diseases such as chronic obstructive pulmonary disease (COPD). However, the pulmonary host defense mechanisms regulating anti-P. aeruginosa immunity remain incompletely understood. Here we demonstrate, by studying an airway P. aeruginosa infection model, in vivo bioluminescence imaging, neutrophil effector responses and human airway samples, that the chemokine receptor CXCR1 regulates pulmonary host defense against P. aeruginosa. Mechanistically, CXCR1 regulates anti-Pseudomonas neutrophil responses through modulation of reactive oxygen species and interference with Toll-like receptor 5 expression. These studies define CXCR1 as a novel, noncanonical chemokine receptor that regulates pulmonary anti-Pseudomonas host defense with broad implications for CF, COPD and other infectious lung diseases.
Collapse
Affiliation(s)
- M Carevic
- Children's Hospital and Interdisciplinary Center for Infectious Diseases, University of Tx00FC;bingen, Tx00FC;bingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
607
|
Alfaro C, Teijeira A, Oñate C, Pérez G, Sanmamed MF, Andueza MP, Alignani D, Labiano S, Azpilikueta A, Rodriguez-Paulete A, Garasa S, Fusco JP, Aznar A, Inogés S, De Pizzol M, Allegretti M, Medina-Echeverz J, Berraondo P, Perez-Gracia JL, Melero I. Tumor-Produced Interleukin-8 Attracts Human Myeloid-Derived Suppressor Cells and Elicits Extrusion of Neutrophil Extracellular Traps (NETs). Clin Cancer Res 2016; 22:3924-36. [PMID: 26957562 DOI: 10.1158/1078-0432.ccr-15-2463] [Citation(s) in RCA: 299] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/03/2016] [Indexed: 12/17/2022]
Abstract
PURPOSE Myeloid-derived suppressor cells (MDSC) are considered an important T-cell immunosuppressive component in cancer-bearing hosts. The factors that attract these cells to the tumor microenvironment are poorly understood. IL8 (CXCL8) is a potent chemotactic factor for neutrophils and monocytes. EXPERIMENTAL DESIGN MDSC were characterized and sorted by multicolor flow cytometry on ficoll-gradient isolated blood leucokytes from healthy volunteers (n = 10) and advanced cancer patients (n = 28). In chemotaxis assays, sorted granulocytic and monocytic MDSC were tested in response to recombinant IL8, IL8 derived from cancer cell lines, and patient sera. Neutrophil extracellular traps (NETs) formation was assessed by confocal microscopy, fluorimetry, and time-lapse fluorescence confocal microscopy on short-term MDSC cultures. RESULTS IL8 chemoattracts both granulocytic (GrMDSC) and monocytic (MoMDSC) human MDSC. Monocytic but not granulocytic MDSC exerted a suppressor activity on the proliferation of autologous T cells isolated from the circulation of cancer patients. IL8 did not modify the T-cell suppressor activity of human MDSC. However, IL8 induced the formation of NETs in the GrMDSC subset. CONCLUSIONS IL8 derived from tumors contributes to the chemotactic recruitment of MDSC and to their functional control. Clin Cancer Res; 22(15); 3924-36. ©2016 AACR.
Collapse
Affiliation(s)
- Carlos Alfaro
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Alvaro Teijeira
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Carmen Oñate
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Guiomar Pérez
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Miguel F Sanmamed
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Maria Pilar Andueza
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Diego Alignani
- Cytometry Platform, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Sara Labiano
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Arantza Azpilikueta
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Alfonso Rodriguez-Paulete
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Saray Garasa
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Juan P Fusco
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Angela Aznar
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Susana Inogés
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | | | | | - Jose Medina-Echeverz
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Pedro Berraondo
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain
| | - Jose L Perez-Gracia
- Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain
| | - Ignacio Melero
- Division of Gene Therapy and Hepatology, Centre for Applied Medical Research (CIMA), Pamplona, Spain. Department of Oncology, University Clinic of Navarra, Pamplona, Spain. Department of Immunology, University Clinic of Navarra, Pamplona, Spain.
| |
Collapse
|
608
|
Rimmelé T, Payen D, Cantaluppi V, Marshall J, Gomez H, Gomez A, Murray P, Kellum JA. IMMUNE CELL PHENOTYPE AND FUNCTION IN SEPSIS. Shock 2016; 45:282-91. [PMID: 26529661 PMCID: PMC4752878 DOI: 10.1097/shk.0000000000000495] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cells of the innate and adaptive immune systems play a critical role in the host response to sepsis. Moreover, their accessibility for sampling and their capacity to respond dynamically to an acute threat increases the possibility that leukocytes might serve as a measure of a systemic state of altered responsiveness in sepsis.The working group of the 14th Acute Dialysis Quality Initiative (ADQI) conference sought to obtain consensus on the characteristic functional and phenotypic changes in cells of the innate and adaptive immune system in the setting of sepsis. Techniques for the study of circulating leukocytes were also reviewed and the impact on cellular phenotypes and leukocyte function of nonextracorporeal treatments and extracorporeal blood purification therapies proposed for sepsis was analyzed.A large number of alterations in the expression of distinct neutrophil and monocyte surface markers have been reported in septic patients. The most consistent alteration seen in septic neutrophils is their activation of a survival program that resists apoptotic death. Reduced expression of HLA-DR is a characteristic finding on septic monocytes, but monocyte antimicrobial function does not appear to be significantly altered in sepsis. Regarding adaptive immunity, sepsis-induced apoptosis leads to lymphopenia in patients with septic shock and it involves all types of T cells (CD4, CD8, and Natural Killer) except T regulatory cells, thus favoring immunosuppression. Finally, numerous promising therapies targeting the host immune response to sepsis are under investigation. These potential treatments can have an effect on the number of immune cells, the proportion of cell subtypes, and the cell function.
Collapse
Affiliation(s)
- Thomas Rimmelé
- Anesthesiology and Critical Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon, University Claude Bernard Lyon 1, Lyon, France
| | - Didier Payen
- Department of Anesthesiology & Critical Care and UMR INSERM 1160; Lariboisière Hospital, AP-HP and University Paris 7, Sorbonne Paris Cité, Paris, France
| | - Vincenzo Cantaluppi
- Nephrology, Dialysis and Kidney Transplantation Unit, Department of Medical Sciences, “Citta' della Salute e della Scienza di Torino- Molinette” University Hospital, Torino, Italy
| | - John Marshall
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario Canada
| | - Hernando Gomez
- Center for Critical Care Nephrology; The CRISMA (Clinical Research, Investigation, and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alonso Gomez
- Academia Colombiana de Medicina Critica (ACOMEC)
- Division of Critical Care Medicine, Clínica Palermo, Bogotá, Colombia
| | | | - John A. Kellum
- Center for Critical Care Nephrology; The CRISMA (Clinical Research, Investigation, and Systems Modeling of Acute Illness) Center, Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
609
|
Abstract
Cystic fibrosis (CF) lung disease is characterized by persistent and unresolved inflammation, with elevated proinflammatory and decreased anti-inflammatory cytokines, and greater numbers of immune cells. Hyperinflammation is recognized as a leading cause of lung tissue destruction in CF. Hyper-inflammation is not solely observed in the lungs of CF patients, since it may contribute to destruction of exocrine pancreas and, likely, to defects in gastrointestinal tract tissue integrity. Paradoxically, despite the robust inflammatory response, and elevated number of immune cells (such as neutrophils and macrophages), CF lungs fail to clear bacteria and are more susceptible to infections. Here, we have summarized the current understanding of immune dysregulation in CF, which may drive hyperinflammation and impaired host defense.
Collapse
Affiliation(s)
- Emanuela M Bruscia
- Section of Respiratory Medicine, Department of Pediatrics, Yale University School of Medicine, 330 Cedar Street, FMP, Room#524, New Haven, CT 06520, USA.
| | - Tracey L Bonfield
- Division of Pulmonology, Allergy and Immunology, Department of Pediatrics, Case Western Reserve University School of Medicine, 0900 Euclid Avenue, Cleveland, OH 44106-4948, USA.
| |
Collapse
|
610
|
Zamakhchari MF, Sima C, Sama K, Fine N, Glogauer M, Van Dyke TE, Gyurko R. Lack of p47(phox) in Akita Diabetic Mice Is Associated with Interstitial Pneumonia, Fibrosis, and Oral Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:659-70. [PMID: 26747235 PMCID: PMC4816692 DOI: 10.1016/j.ajpath.2015.10.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 10/06/2015] [Accepted: 10/29/2015] [Indexed: 01/22/2023]
Abstract
Excess reactive oxygen species production is central to the development of diabetic complications. The contribution of leukocyte reactive oxygen species produced by the NADPH oxidase to altered inflammatory responses associated with uncontrolled hyperglycemia is poorly understood. To get insight into the role of phagocytic superoxide in the onset of diabetic complications, we used a model of periodontitis in mice with chronic hyperglycemia and lack of leukocyte p47(phox) (Akita/Ncf1) bred from C57BL/6-Ins2(Akita)/J (Akita) and neutrophil cytosolic factor 1 knockout (Ncf1) mice. Akita/Nfc1 mice showed progressive cachexia starting at early age and increased mortality by six months. Their lungs developed infiltrative interstitial lesions that obliterated air spaces as early as 12 weeks when fungal colonization of lungs also was observed. Neutrophils of Akita/Ncf1 mice had normal degranulation and phagocytic efficiency when compared with wild-type mice. Although Akita/Ncf1 mice had increased prevalence of oral infections and more severe periodontitis compared with wild-type mice, bone loss was only marginally higher compared with Akita and Ncf1 null mice. Altogether these results indicate that lack of leukocyte superoxide production in mice with chronic hyperglycemia results in interstitial pneumonia and increased susceptibility to infections.
Collapse
Affiliation(s)
- Mai F Zamakhchari
- Department of Periodontology and Oral Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts
| | - Corneliu Sima
- Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, Massachusetts; Department of Oral Medicine, Infection, Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Kishore Sama
- Department of Periodontology and Oral Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts
| | - Noah Fine
- The Matrix Dynamics Group, University of Toronto, Toronto, Ontario, Canada
| | - Michael Glogauer
- The Matrix Dynamics Group, University of Toronto, Toronto, Ontario, Canada
| | - Thomas E Van Dyke
- Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, Massachusetts; Department of Oral Medicine, Infection, Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Robert Gyurko
- Department of Periodontology and Oral Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts; Department of Periodontology, Tufts University School of Dental Medicine, Boston, Massachusetts.
| |
Collapse
|
611
|
Kaufmann SH, Dorhoi A. Molecular Determinants in Phagocyte-Bacteria Interactions. Immunity 2016; 44:476-491. [DOI: 10.1016/j.immuni.2016.02.014] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 01/28/2016] [Accepted: 02/17/2016] [Indexed: 12/24/2022]
|
612
|
Montserrat-de la Paz S, Fernandez-Arche A, de la Puerta R, Quilez AM, Muriana FJG, Garcia-Gimenez MD, Bermudez B. Mitraphylline inhibits lipopolysaccharide-mediated activation of primary human neutrophils. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:141-148. [PMID: 26926175 DOI: 10.1016/j.phymed.2015.12.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/17/2015] [Accepted: 12/21/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Mitraphylline (MTP) is the major pentacyclic oxindolic alkaloid presented in Uncaria tomentosa. It has traditionally been used to treat disorders including arthritis, heart disease, cancer, and other inflammatory diseases. However, the specific role of MTP is still not clear, with more comprehensivestudies, our understanding of this ancient herbal medicine will continue growing. HYPOTHESIS/PURPOSE Some studies provided its ability to inhibit proinflamatory cytokines, such as TNF-α, through NF-κB-dependent mechanism. TNF-α primes neutrophils and modulates phagocytic and oxidative burst activities in inflammatory processes. Since, neutrophils represent the most abundant pool of leukocytes in human blood and play a crucial role in inflammation, we aimed to determine the ability of MTP to modulate neutrophil activation and differentially regulate inflammatory-related cytokines. METHODS To determine the mechanism of action of MTP, we investigated the effects on LPS-activated human primary neutrophils responses including activation surface markers by FACS and the expression of inflammatory cytokines, measured by real time PCR and ELISA. RESULTS Treatment with MTP reduced the LPS-dependent activation effects. Activated neutrophils (CD16(+)CD62L(-)) diminished after MTP administration. Moreover, proinflamatory cytokines (TNF-α, IL-6 or IL-8) expression and secretion were concomitantly reduced, similar to basal control conditions. CONCLUSION Taken together, our results demonstrate that MTP is able to elicit an anti-inflammatory response that modulates neutrophil activation contributing to the attenuation of inflammatory episodes. Further studies are need to characterize the mechanism by which MTP can affect this pathway that could provide a means to develop MTP as new candidate for inflammatory disease therapies.
Collapse
Affiliation(s)
- Sergio Montserrat-de la Paz
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012, Seville, Spain ; Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, Seville, Spain
| | - Angeles Fernandez-Arche
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012, Seville, Spain
| | - Rocío de la Puerta
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012, Seville, Spain
| | - Ana M Quilez
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012, Seville, Spain
| | - Francisco J G Muriana
- Laboratory of Cellular and Molecular Nutrition, Instituto de la Grasa, CSIC, Seville, Spain
| | | | - Beatriz Bermudez
- Department of Pharmacology, School of Pharmacy, University of Seville, 41012, Seville, Spain .
| |
Collapse
|
613
|
Endothelial CD36 Contributes to Postischemic Brain Injury by Promoting Neutrophil Activation via CSF3. J Neurosci 2016; 35:14783-93. [PMID: 26538649 DOI: 10.1523/jneurosci.2980-15.2015] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
UNLABELLED The scavenger receptor CD36 is a critical factor initiating ischemic brain injury, but the cell type(s) expressing CD36 and responsible for its harmful effects remain unknown. Using bone marrow (BM) chimeras subjected to transient middle cerebral artery occlusion, we found that CD36(-/-) mice transplanted with wild-type (WT) BM (WT→CD36(-/-)) have smaller infarcts (-67%), comparable with those of mice lacking CD36 both in brain and hematogenous cells (CD36(-/-) →CD36(-/-); - 72%). Conversely, WT mice receiving CD36(-/-) BM (CD36(-/-) →WT) have infarcts similar to WT→WT mice, suggesting that CD36 in the host brain (i.e., in microglia and endothelial cells), and not in hematogenous cells is involved in the damage. As anticipated, postischemic neutrophil infiltration in CD36(-/-) →CD36(-/-) mice was attenuated. Surprisingly, however, in WT→CD36(-/-) mice, in which infarcts were small, neutrophil infiltration was large and similar to that of CD36(-/-) →WT mice, in which infarcts were not reduced. Postischemic neutrophil free radical production was attenuated in WT→CD36(-/-) mice compared with CD36(-/-) →WT mice, whereas expression of the neutrophil activator colony-stimulating factor 3 (CSF3) was suppressed in CD36(-/-) cerebral endothelial cells, but not microglia. In CD36(-/-) cerebral endothelial cultures exposed to extracts from stroke brains, the upregulation of CSF3, but not neutrophil attractant chemokines, was suppressed. Intracerebroventricular administration of CSF3, 24 h after stroke, reconstituted neutrophil radical production and increased infarct volume in WT→CD36(-/-) mice. The findings identify endothelial cells as a key player in the deleterious effects of CD36 in stroke, and unveil a novel role of endothelial CD36 in enabling neutrophil neurotoxicity through CSF3. SIGNIFICANCE STATEMENT Ischemic stroke is a leading cause of death and disability worldwide with limited therapeutic options. The inflammatory response initiated by cerebral ischemia-reperfusion contributes to ischemic brain injury and is a potential therapeutic target. Here we report that CD36, an innate immunity receptor involved in the initiation of postischemic inflammation, is a previously unrecognized regulator of neutrophil cytotoxicity. The effect is mediated by endothelial CD36 via upregulation of the neutrophil activator CSF3 in cerebral endothelial cells. Therefore, approaches to modulate cerebral endothelial CD36 signaling or to neutralize CSF3 may provide novel therapeutic opportunities to ameliorate postischemic inflammatory injury.
Collapse
|
614
|
Engert A, Balduini C, Brand A, Coiffier B, Cordonnier C, Döhner H, de Wit TD, Eichinger S, Fibbe W, Green T, de Haas F, Iolascon A, Jaffredo T, Rodeghiero F, Salles G, Schuringa JJ. The European Hematology Association Roadmap for European Hematology Research: a consensus document. Haematologica 2016; 101:115-208. [PMID: 26819058 PMCID: PMC4938336 DOI: 10.3324/haematol.2015.136739] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/27/2016] [Indexed: 01/28/2023] Open
Abstract
The European Hematology Association (EHA) Roadmap for European Hematology Research highlights major achievements in diagnosis and treatment of blood disorders and identifies the greatest unmet clinical and scientific needs in those areas to enable better funded, more focused European hematology research. Initiated by the EHA, around 300 experts contributed to the consensus document, which will help European policy makers, research funders, research organizations, researchers, and patient groups make better informed decisions on hematology research. It also aims to raise public awareness of the burden of blood disorders on European society, which purely in economic terms is estimated at €23 billion per year, a level of cost that is not matched in current European hematology research funding. In recent decades, hematology research has improved our fundamental understanding of the biology of blood disorders, and has improved diagnostics and treatments, sometimes in revolutionary ways. This progress highlights the potential of focused basic research programs such as this EHA Roadmap.The EHA Roadmap identifies nine 'sections' in hematology: normal hematopoiesis, malignant lymphoid and myeloid diseases, anemias and related diseases, platelet disorders, blood coagulation and hemostatic disorders, transfusion medicine, infections in hematology, and hematopoietic stem cell transplantation. These sections span 60 smaller groups of diseases or disorders.The EHA Roadmap identifies priorities and needs across the field of hematology, including those to develop targeted therapies based on genomic profiling and chemical biology, to eradicate minimal residual malignant disease, and to develop cellular immunotherapies, combination treatments, gene therapies, hematopoietic stem cell treatments, and treatments that are better tolerated by elderly patients.
Collapse
Affiliation(s)
| | | | - Anneke Brand
- Leids Universitair Medisch Centrum, Leiden, the Netherlands
| | | | | | | | | | | | - Willem Fibbe
- Leids Universitair Medisch Centrum, Leiden, the Netherlands
| | - Tony Green
- Cambridge Institute for Medical Research, United Kingdom
| | - Fleur de Haas
- European Hematology Association, The Hague, the Netherlands
| | | | | | | | - Gilles Salles
- Hospices Civils de Lyon/Université de Lyon, Pierre-Bénite, France
| | | |
Collapse
|
615
|
Marcinkiewicz J, Stręk P, Strus M, Głowacki R, Ciszek-Lenda M, Zagórska-Świeży K, Gawda A, Tomusiak A. Staphylococcus epidermidis and biofilm-associated neutrophils in chronic rhinosinusitis. A pilot study. Int J Exp Pathol 2016; 96:378-86. [PMID: 26765504 DOI: 10.1111/iep.12156] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 09/16/2015] [Indexed: 11/26/2022] Open
Abstract
A key role of bacterial biofilm in the pathogenesis of chronic rhinosinusitis (CRS) with (CRSwNP) and without nasal polyps (CRSsNP) is commonly accepted. However, the impact of some bacterial species isolated from inflamed sinus mucosa on biofilm formation is unclear. In particular, the role of Staphylococcus epidermidis as aetiological agents of CRS is controversial. Moreover, the effect of biofilm formation on neutrophil infiltration and activity in CRSwNP calls for explanation. In this study, biofilms were found in three of 10 patients (mean age = 46 ± 14) with CRS undergoing endoscopic sinus surgery by means of scanning electron microscopy. Unexpectedly, S. epidermidis was the primary isolated bacteria and was also found to be present in all biofilm-positive mucosa specimens, indicating its pivotal role in the pathogenesis of severe chronic infections associated with biofilm formation. We have also measured the activity of myeloperoxidase (MPO), the most abundant neutrophil enzyme, to demonstrate the presence of neutrophils in the samples tested. Our present results show that the level of MPO in CRS associated with biofilm is lower than that without biofilm. It may suggest either a low number of neutrophils or the presence of a type of neutrophils with compromised antimicrobial activity, described as biofilm-associated neutrophils (BAN). Finally, we conclude that further studies with a large number of CRS cases should be performed to establish the association between S. epidermidis and other frequently isolated bacterial species from paranasal sinuses, with the severity of CRS, biofilm formation and the infiltration of BAN.
Collapse
Affiliation(s)
| | - Paweł Stręk
- Department of Otorhinolaryngology, Jagiellonian University Medical College, Krakow, Poland
| | - Magdalena Strus
- Chair of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| | - Roman Głowacki
- Department of Otorhinolaryngology, Jagiellonian University Medical College, Krakow, Poland
| | - Marta Ciszek-Lenda
- Chair of Immunology, Jagiellonian University Medical College, Krakow, Poland
| | | | - Anna Gawda
- Chair of Immunology, Jagiellonian University Medical College, Krakow, Poland
| | - Anna Tomusiak
- Chair of Microbiology, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
616
|
Wichert S, Pettersson Å, Hellmark T, Johansson Å, Hansson M. Phagocyte function decreases after high-dose treatment with melphalan and autologous stem cell transplantation in patients with multiple myeloma. Exp Hematol 2016; 44:342-351.e5. [PMID: 26774385 DOI: 10.1016/j.exphem.2016.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 12/19/2015] [Accepted: 01/06/2016] [Indexed: 11/28/2022]
Abstract
High-dose melphalan with autologous hematopoietic stem cell transplantation (ASCT) is the standard of care for younger patients with newly diagnosed multiple myeloma and is aimed at achieving as deep and complete a response as possible after various combinations of induction therapy. However, it is frequently associated with infectious complications. This study investigated the effects of high-dose treatment with autologous stem cell support on patients' innate immunity, with a focus on subpopulations and functioning of recently released polymorphonuclear leukocytes (PMNs) and monocytes in peripheral blood. Flow cytometry-based analysis was used to measure the degree of PMN maturation and activation, before and after ASCT and compared with healthy controls. After high-dose treatment and ASCT, a smaller proportion of patients' PMNs had the capacity for oxidative burst. Moreover, patients' PMNs, both before and after ASCT, had a reduced capacity for phagocytosis. Eosinophils, which recently have been suggested to play a role in promoting malignant plasma cell proliferation, were markedly reduced after ASCT, with slow regeneration. HLA-DR expression by monocytes was significantly depressed after ASCT, a characteristic often attributed to monocytic myeloid-derived suppressor cells. Our results suggest that several aspects of phagocytic function are impaired for at least 20 days after ASCT.
Collapse
Affiliation(s)
- Stina Wichert
- Department of Hematology, Skåne University Hospital and Lund University, Lund, Sweden.
| | - Åsa Pettersson
- Department of Nephrology, Clinical Sciences in Lund, Lund University, Lund, Sweden
| | - Thomas Hellmark
- Department of Nephrology, Clinical Sciences in Lund, Lund University, Lund, Sweden
| | - Åsa Johansson
- Department of Hematology, Skåne University Hospital and Lund University, Lund, Sweden; Clinical Immunology and Transfusion Medicine, University and Regional Laboratories Region Skåne, Lund, Sweden
| | - Markus Hansson
- Department of Hematology, Skåne University Hospital and Lund University, Lund, Sweden
| |
Collapse
|
617
|
Thymoquinone strongly inhibits fMLF-induced neutrophil functions and exhibits anti-inflammatory properties in vivo. Biochem Pharmacol 2016; 104:62-73. [PMID: 26774451 DOI: 10.1016/j.bcp.2016.01.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/06/2016] [Indexed: 11/21/2022]
Abstract
Polymorphonuclear neutrophils are key players in host defense against pathogens through the robust production of superoxide anion by the NADPH oxidase and the release of antibacterial proteins from granules. However, inappropriate release of these agents in the extracellular environment induces severe tissue injury, thereby contributing to the physiopathology of acute and chronic inflammatory disorders. Many studies have been carried out to identify molecules capable of inhibiting phagocyte functions, in particular superoxide anion production, for therapeutic purposes. In the present study, we show that thymoquinone (TQ), the major component of the volatile oil from Nigella sativa (black cumin) seeds strongly inhibits fMLF-induced superoxide production and granules exocytosis in neutrophils. The inhibition of superoxide anion was not due to a scavenger effect, as TQ did not inhibit superoxide anion produced by the xanthine/xanthine oxidase system. Interestingly, TQ impaired the phosphorylation on Ser-304 and Ser-328 of p47(PHOX), a cytosolic subunit of the NADPH oxidase. TQ also attenuated specific and azurophilic granule exocytosis in fMLF-stimulated neutrophils as evidenced by decreased cell surface expression of gp91(PHOX) and CD11b, and release of myeloperoxidase. Furthermore, both the PKC and MAPK pathways, which are involved in p47(PHOX) phosphorylation and granules exocytosis, respectively, were inhibited by TQ in fMLF-stimulated neutrophils. Finally, in a model of pleurisy induced by λ-carrageenan in rats, TQ reduced neutrophil accumulation in the pleural space, showing that it not only inhibits PMN functions in vitro, but also exhibits anti-inflammatory properties in vivo. Thus, TQ possesses promising anti-inflammatory therapeutic potential.
Collapse
|
618
|
Neutrophils, platelets, and inflammatory pathways at the nexus of sickle cell disease pathophysiology. Blood 2016; 127:801-9. [PMID: 26758915 DOI: 10.1182/blood-2015-09-618538] [Citation(s) in RCA: 292] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 10/28/2015] [Indexed: 02/07/2023] Open
Abstract
Sickle cell disease (SCD) is a severe genetic blood disorder characterized by hemolytic anemia, episodic vaso-occlusion, and progressive organ damage. Current management of the disease remains symptomatic or preventative. Specific treatment targeting major complications such as vaso-occlusion is still lacking. Recent studies have identified various cellular and molecular factors that contribute to the pathophysiology of SCD. Here, we review the role of these elements and discuss the opportunities for therapeutic intervention.
Collapse
|
619
|
Abstract
West Nile virus (WNV) can cause severe neuroinvasive disease in humans and currently no vaccine or specific treatments are available. As aging is the most prominent risk factor for WNV, age-related immune dysregulation likely plays an essential role in host susceptibility to infection with WNV. In this review, we summarize recent findings in effects of aging on immune responses to WNV infection. In particular, we focus on the age-dependent dysregulation of innate immune cell types-neutrophils, macrophages, and dendritic cells-in response to WNV infection, as well as age-related alterations in NK cells and γδ T cells that may associate with increased WNV susceptibility in older people. We also highlight two advanced technologies, i.e., mass cytometry and microRNA profiling, which significantly contribute to systems-level study of immune dysregulation in aging and should facilitate new discoveries for therapeutic intervention against WNV.
Collapse
Affiliation(s)
- Yi Yao
- Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA
| | - Ruth R Montgomery
- Department of Internal Medicine, Yale University School of Medicine, 300 Cedar Street, New Haven, CT, 06520, USA.
| |
Collapse
|
620
|
|
621
|
Alba G, Reyes-Quiróz ME, Sáenz J, Geniz I, Jiménez J, Martín-Nieto J, Pintado E, Sobrino F, Santa-María C. 7-Keto-cholesterol and 25-hydroxy-1 cholesterol rapidly enhance ROS production in human neutrophils. Eur J Nutr 2015; 55:2485-2492. [DOI: 10.1007/s00394-015-1142-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 12/15/2015] [Indexed: 10/22/2022]
|
622
|
Baruah S, Keck K, Vrenios M, Pope MR, Pearl M, Doerschug K, Klesney-Tait J. Identification of a Novel Splice Variant Isoform of TREM-1 in Human Neutrophil Granules. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:5725-31. [PMID: 26561551 PMCID: PMC4670805 DOI: 10.4049/jimmunol.1402713] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 10/09/2015] [Indexed: 01/28/2023]
Abstract
Triggering receptor expressed on myeloid cells-1 (TREM-1) is critical for inflammatory signal amplification. Humans have two forms of TREM-1: a membrane receptor, associated with the adaptor DAP12, and a soluble receptor detected at times of infection. The membrane receptor isoform acts synergistically with the TLR pathway to promote cytokine secretion and neutrophil migration, whereas the soluble receptor functions as a counterregulatory molecule. In multiple models of sepsis, exogenous administration of soluble forms of TREM-1 attenuates inflammation and markedly improves survival. Despite intense interest in soluble TREM-1, both as a clinical predictor of survival and as a therapeutic tool, the origin of native soluble TREM-1 remains controversial. Using human neutrophils, we identified a 15-kDa TREM-1 isoform in primary (azurophilic) and secondary (specific) granules. Mass spectrometric analysis, ELISA, and immunoblot confirm that the 15-kDa protein is a novel splice variant form of TREM-1 (TREM-1sv). Neutrophil stimulation with Pseudomonas aeruginosa, LPS, or PAM(3)Cys4 resulted in degranulation and release of TREM-1sv. The addition of exogenous TREM-1sv inhibited TREM-1 receptor-mediated proinflammatory cytokine production. Thus, these data reveal that TREM-1 isoforms simultaneously activate and inhibit inflammation via the canonical membrane TREM-1 molecule and this newly discovered granular isoform, TREM-1sv.
Collapse
Affiliation(s)
- Sankar Baruah
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Kathy Keck
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Michelle Vrenios
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Marshall R Pope
- Carver College of Medicine Proteomics Facility, University of Iowa, Iowa City, IA 52242; and
| | | | - Kevin Doerschug
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Julia Klesney-Tait
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242;
| |
Collapse
|
623
|
Anti-inflammatory effects of Perilla frutescens in activated human neutrophils through two independent pathways: Src family kinases and Calcium. Sci Rep 2015; 5:18204. [PMID: 26659126 PMCID: PMC4677386 DOI: 10.1038/srep18204] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 10/07/2015] [Indexed: 11/12/2022] Open
Abstract
The leaves of Perilla frutescens (L.) Britt. have been traditionally used as an herbal medicine in East Asian countries to treat a variety diseases. In this present study, we investigated the inhibitory effects of P. frutescens extract (PFE) on N-formyl-Met-Leu-Phe (fMLF)-stimulated human neutrophils and the underlying mechanisms. PFE (1, 3, and 10 μg/ml) inhibited superoxide anion production, elastase release, reactive oxygen species formation, CD11b expression, and cell migration in fMLF-activated human neutrophils in dose-dependent manners. PFE inhibited fMLF-induced phosphorylation of the Src family kinases (SFKs), Src (Tyr416) and Lyn (Tyr396), and reduced their enzymatic activities. Both PFE and PP2 (a selective inhibitor of SFKs) reduced the phosphorylation of Burton’s tyrosine kinases (Tyr223) and Vav (Tyr174) in fMLF-activated human neutrophils. Additionally, PFE decreased intracellular Ca2+ levels ([Ca2+]i), whereas PP2 prolonged the time required for [Ca2+]i to return to its basal level. Our findings indicated that PFE effectively regulated the inflammatory activities of fMLF-activated human neutrophils. The anti-inflammatory effects of PFE on activated human neutrophils were mediated through two independent signaling pathways involving SFKs (Src and Lyn) and mobilization of intracellular Ca2+.
Collapse
|
624
|
Powell DR, Huttenlocher A. Neutrophils in the Tumor Microenvironment. Trends Immunol 2015; 37:41-52. [PMID: 26700397 DOI: 10.1016/j.it.2015.11.008] [Citation(s) in RCA: 436] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 01/13/2023]
Abstract
Neutrophils are the first responders to sites of acute tissue damage and infection. Recent studies suggest that in addition to neutrophil apoptosis, resolution of neutrophil inflammation at wounds can be mediated by reverse migration from tissues and transmigration back into the vasculature. In settings of chronic inflammation, neutrophils persist in tissues, and this persistence has been associated with cancer progression. However, the role of neutrophils in the tumor microenvironment remains controversial, with evidence for both pro- and anti-tumor roles. Here we review the mechanisms that regulate neutrophil recruitment and resolution at sites of tissue damage, with a specific focus on the tumor microenvironment. We discuss the current understanding as to how neutrophils alter the tumor microenvironment to support or hinder cancer progression, and in this context outline gaps in understanding and important areas of inquiry.
Collapse
Affiliation(s)
- Davalyn R Powell
- Departments of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Anna Huttenlocher
- Departments of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
625
|
Crupi A, Costa A, Tarnok A, Melzer S, Teodori L. Inflammation in tissue engineering: The Janus between engraftment and rejection. Eur J Immunol 2015; 45:3222-36. [DOI: 10.1002/eji.201545818] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 10/07/2015] [Accepted: 11/05/2015] [Indexed: 01/10/2023]
Affiliation(s)
- Annunziata Crupi
- Department of Fusion and Technologies for Nuclear Safety and Security; Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA; Frascati-Rome Italy
- Fondazione San Raffaele; Ceglie Messapica Italy
| | - Alessandra Costa
- Department of Surgery; McGowan Institute; University of Pittsburgh Medical Center; Pittsburgh PA USA
| | - Attila Tarnok
- Department of Pediatric Cardiology; Heart Center GmbH Leipzig; and Translational Center for Regenerative Medicine; University Leipzig; Leipzig Germany
| | - Susanne Melzer
- Department of Pediatric Cardiology; Heart Center GmbH Leipzig; and Translational Center for Regenerative Medicine; University Leipzig; Leipzig Germany
| | - Laura Teodori
- Department of Fusion and Technologies for Nuclear Safety and Security; Diagnostic and Metrology (FSN-TECFIS-DIM), ENEA; Frascati-Rome Italy
- Fondazione San Raffaele; Ceglie Messapica Italy
| |
Collapse
|
626
|
Björnsdottir H, Welin A, Michaëlsson E, Osla V, Berg S, Christenson K, Sundqvist M, Dahlgren C, Karlsson A, Bylund J. Neutrophil NET formation is regulated from the inside by myeloperoxidase-processed reactive oxygen species. Free Radic Biol Med 2015; 89:1024-35. [PMID: 26459032 DOI: 10.1016/j.freeradbiomed.2015.10.398] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/11/2015] [Accepted: 10/07/2015] [Indexed: 11/21/2022]
Abstract
AIM Neutrophil extracellular traps (NETs) are mesh-like DNA fibers clad with intracellular proteins that are cast out from neutrophils in response to certain stimuli. The process is thought to depend on reactive oxygen species (ROS) generated by the phagocyte NADPH-oxidase and the ROS-modulating granule enzyme myeloperoxidase (MPO), but when, how, and where these factors contribute is so far uncertain. The neutrophil NADPH-oxidase can be activated at different cellular sites and ROS may be produced and processed by MPO within intracellular granules, even in situations where a phagosome is not formed, e.g., upon stimulation with phorbol myristate acetate (PMA). OBJECTIVES We investigated the subcellular location of ROS production and processing by MPO in the context of PMA-induced NET formation. RESULTS Complete neutralization of extracellular ROS was not sufficient to block NET formation triggered by PMA, indicating that intragranular ROS are critical for NETosis. Employing a set of novel MPO-inhibitors, inhibition of NET formation correlated with inhibition of intragranular MPO activity. Also, extracellular addition of MPO was not sufficient to rescue NET formation in completely MPO-deficient neutrophils and specific neutralization by luminol of MPO-processed ROS within intracellular granules led to a complete block of PMA-triggered NET formation. CONCLUSION We show for the first time that inhibition of intragranular MPO activity, or neutralization of intragranular MPO-processed ROS by luminol effectively block NET formation. Our data demonstrate that ROS must be formed and processed by MPO in order to trigger NET formation, and that these events have to occur within intracellular granules.
Collapse
Affiliation(s)
- Halla Björnsdottir
- The Phagocyte Research Group, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Amanda Welin
- The Phagocyte Research Group, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Erik Michaëlsson
- Department of Bioscience, CVMD iMed, AstraZeneca R&D Mölndal, Mölndal, Sweden
| | - Veronica Osla
- The Phagocyte Research Group, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Stefan Berg
- Department of Pediatrics, The Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Karin Christenson
- The Phagocyte Research Group, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Martina Sundqvist
- The Phagocyte Research Group, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Claes Dahlgren
- The Phagocyte Research Group, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anna Karlsson
- The Phagocyte Research Group, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Johan Bylund
- The Phagocyte Research Group, Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Department of Oral Microbiology and Immunology, Institute of Odontology, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
627
|
Ueda R, Narumi K, Hashimoto H, Miyakawa R, Okusaka T, Aoki K. Interaction of natural killer cells with neutrophils exerts a significant antitumor immunity in hematopoietic stem cell transplantation recipients. Cancer Med 2015; 5:49-60. [PMID: 26589884 PMCID: PMC4708905 DOI: 10.1002/cam4.550] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/27/2015] [Accepted: 09/01/2015] [Indexed: 12/30/2022] Open
Abstract
Autologous hematopoietic stem cell transplantation (HSCT) can induce a strong antitumor immunity by homeostatic proliferation (HP) of T cells and suppression of regulatory T cells following preconditioning‐induced lymphopenia. However, the role of innate immunity including natural killer (NK) cells is still not understood. Here, first, we examined whether NK cells exert an antitumor effect after syngeneic HSCT in a murine colon cancer model. Flow cytometry showed that NK cells as well as T cells rapidly proliferated after HSCT, and the frequency of mature NK cells was increased in tumor during HP. Furthermore, NK cells undergoing HP were highly activated, which contributed to substantial tumor suppression. Then, we found that a large number of neutrophils accumulated in tumor early after syngeneic HSCT. It was recently reported that neutrophil‐derived mediators modulate NK cell effector functions, and so we examined whether the neutrophils infiltrated in tumor are associated with NK cell‐mediated antitumor effect. The depletion of neutrophils significantly impaired an activation of NK cells in tumor and increased the fraction of proliferative NK cells accompanied by a decrease in NK cell survival. The results suggested that neutrophils in tumor prevent NK cells from activation‐induced cell death during HP, thus leading to a significant antitumor effect by NK cells. This study revealed a novel aspect of antitumor immunity induced by HSCT and may contribute to the development of an effective therapeutic strategy for cancer using HSCT.
Collapse
Affiliation(s)
- Ryosuke Ueda
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kenta Narumi
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Hisayoshi Hashimoto
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Reina Miyakawa
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Takuji Okusaka
- Division of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Kazunori Aoki
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| |
Collapse
|
628
|
A novel mechanism for NETosis provides antimicrobial defense at the oral mucosa. Blood 2015; 126:2128-37. [DOI: 10.1182/blood-2015-04-641142] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 07/20/2015] [Indexed: 12/18/2022] Open
Abstract
Key Points
Saliva induces bactericidal and DNase resistant NETs in the oral cavity via sialyl LewisX- L-selectin signaling. Disordered homeostasis in the oral cavity may lead to deficient saliva-mediated NETosis.
Collapse
|
629
|
Menzel LP, Bigger CH. Identification of unstimulated constitutive immunocytes, by enzyme histochemistry, in the coenenchyme of the octocoral Swiftia exserta. THE BIOLOGICAL BULLETIN 2015; 229:199-208. [PMID: 26504160 DOI: 10.1086/bblv229n2p199] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Most animals rely on circulating hemocytes as cellular effectors of immunity. These cells traditionally have been characterized by morphology, function, and cellular contents. Morphological descriptions use granule differences and cell shapes; functional descriptions rely on phagocytic ability and oxygen transport; and cellular content descriptions include cytochemical features and key enzymes. Key enzymes used to identify phagocytes in tissues include hydrolytic enzymes, peroxidase, and--in invertebrates--phenoloxidase. Cnidaria such as Swiftia exserta lack a circulatory system, thereby complicating the identification of immune effector cells. As a first step in identifying immunocytes, this study focused on basic enzymes used during phagocytosis and encapsulation; both processes have been reported in octocorals such as S. exserta. Earlier work suggested that there are two populations of phagocytic cells: a constitutive population and an induced population following a trauma-associated challenge. To identify the constitutive immune effector cells in S. exserta in a nonactivated state, we used cryosections of unstimulated animals and the following enzymes to serve as identifying proxies due to their roles in phagocytosis and encapsulation: (1) acid phosphatase, (2) alkaline phosphatase, (3) non-specific esterase, (4) β-glucuronidase, (5) peroxidase, and (6) phenoloxidase. Our results indicate that in unstimulated animals, two distinct cell populations could function as immunocytes. These cell types were differentiated by their enzyme reactivity and their location within the mesoglea of S. exserta, and have been described as either "oblong granular cells" or "granular amoebocytes."
Collapse
Affiliation(s)
- Lorenzo P Menzel
- Department of Biological Sciences, Florida International University, 11200 S.W. 8 Street, Miami, Florida 33199
| | - Charles H Bigger
- Department of Biological Sciences, Florida International University, 11200 S.W. 8 Street, Miami, Florida 33199
| |
Collapse
|
630
|
Antonioli L, Colucci R, Pellegrini C, Giustarini G, Sacco D, Tirotta E, Caputi V, Marsilio I, Giron MC, Németh ZH, Blandizzi C, Fornai M. The AMPK enzyme-complex: from the regulation of cellular energy homeostasis to a possible new molecular target in the management of chronic inflammatory disorders. Expert Opin Ther Targets 2015; 20:179-91. [DOI: 10.1517/14728222.2016.1086752] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
631
|
Lämmermann T. In the eye of the neutrophil swarm-navigation signals that bring neutrophils together in inflamed and infected tissues. J Leukoc Biol 2015; 100:55-63. [PMID: 26416718 DOI: 10.1189/jlb.1mr0915-403] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 09/08/2015] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are sentinel cells that express in higher vertebrates >30 chemokine and chemoattractant receptors to sense and quickly react to tissue damage signals. Intravital microscopy studies in mouse models of wounding, inflammation, and infection have revealed that neutrophils form cell swarms at local sites of tissue injury and cell death. This swarming response is choreographed by chemokines, lipids, and other chemoattractants, controlling sequential phases of highly coordinated chemotaxis, intercellular signal relay, and cluster formation among neutrophils. This review will give a brief overview about the basic principles and key molecules that have led to the refined multistep model of how neutrophils come together to isolate sites of tissue injury and microbial invasion from healthy tissue. Whereas auto- and paracrine signaling among neutrophils during later phases of swarming can provide a level of self-organization for robust navigation in diverse inflammatory settings, guidance factors from primary tissue lesions, resident bystander cells, and dying cells regulate the initial phases of the swarming response. This review will discuss how the specific environmental context and mixture of attractants at the locally inflamed site can lead to variants of the multistep attraction model and influence the extent of neutrophil swarming, ranging from accumulations of only few individual cells to the aggregation of several hundreds of neutrophils, as found in abscesses. Given the critical roles of neutrophils in both host protection and tissue destruction, novel insights on neutrophil swarming might provide useful for the therapeutic modulation of neutrophil-dependent inflammatory processes.
Collapse
Affiliation(s)
- Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| |
Collapse
|
632
|
Bishop CV, Xu F, Molskness TA, Stouffer RL, Hennebold JD. Dynamics of Immune Cell Types Within the Macaque Corpus Luteum During the Menstrual Cycle: Role of Progesterone. Biol Reprod 2015; 93:112. [PMID: 26400401 DOI: 10.1095/biolreprod.115.132753] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 09/18/2015] [Indexed: 12/20/2022] Open
Abstract
The goal of the current study was to characterize the immune cell types within the primate corpus luteum (CL). Luteal tissue was collected from rhesus females at discrete intervals during the luteal phase of the natural menstrual cycle. Dispersed cells were incubated with fluorescently labeled antibodies specific for the immune cell surface proteins CD11b (neutrophils and monocytes/macrophages), CD14 (monocytes/macrophages), CD16 (natural killer [NK] cells), CD20 (B-lymphocytes), and CD3epsilon (T-lymphocytes) for analysis by flow cytometry. Numbers of CD11b-positive (CD11b(+)) and CD14(+) cells increased significantly 3 to 4 days after serum progesterone (P4) concentrations declined below 0.3 ng/ml. CD16(+) cells were the most abundant immune cell type in CL during the mid and mid-late luteal phases and were 3-fold increased 3 to 4 days after serum P4 decreased to baseline levels. CD3epsilon(+) cells tended to increase 3 to 4 days after P4 decline. To determine whether immune cells were upregulated by the loss of luteotropic (LH) support or through loss of LH-dependent steroid milieu, monkeys were assigned to 4 groups: control (no treatment), the GnRH antagonist Antide, Antide plus synthetic progestin (R5020), or Antide plus the estrogen receptor agonists diarylpropionitrile (DPN)/propyl-pyrazole-triol (PPT) during the mid-late luteal phase. Antide treatment increased the numbers of CD11b(+) and CD14(+) cells, whereas progestin, but not estrogen, replacement suppressed the numbers of CD11b(+), CD14(+), and CD16(+) cells. Neither Antide nor steroid replacement altered numbers of CD3epsilon(+) cells. These data suggest that increased numbers of innate immune cells in primate CL after P4 synthesis declines play a role in onset of structural regression of primate CL.
Collapse
Affiliation(s)
- Cecily V Bishop
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon
| | - Fuhua Xu
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon
| | - Theodore A Molskness
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon
| | - Richard L Stouffer
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon Division of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon
| | - Jon D Hennebold
- Division of Reproductive and Developmental Sciences, Oregon Health & Science University, Portland, Oregon Division of Obstetrics and Gynecology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
633
|
Zhang D, Chen G, Manwani D, Mortha A, Xu C, Faith JJ, Burk RD, Kunisaki Y, Jang JE, Scheiermann C, Merad M, Frenette PS. Neutrophil ageing is regulated by the microbiome. Nature 2015; 525:528-32. [PMID: 26374999 PMCID: PMC4712631 DOI: 10.1038/nature15367] [Citation(s) in RCA: 602] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/29/2015] [Indexed: 12/11/2022]
Abstract
Blood polymorphonuclear neutrophils provide immune protection against pathogens, but may also promote tissue injury in inflammatory diseases. Although neutrophils are generally considered to be a relatively homogeneous population, evidence for heterogeneity is emerging. Under steady-state conditions, neutrophil heterogeneity may arise from ageing and replenishment by newly released neutrophils from the bone marrow. Aged neutrophils upregulate CXCR4, a receptor allowing their clearance in the bone marrow, with feedback inhibition of neutrophil production via the IL-17/G-CSF axis, and rhythmic modulation of the haematopoietic stem-cell niche. The aged subset also expresses low levels of L-selectin. Previous studies have suggested that in vitro-aged neutrophils exhibit impaired migration and reduced pro-inflammatory properties. Here, using in vivo ageing analyses in mice, we show that neutrophil pro-inflammatory activity correlates positively with their ageing whilst in circulation. Aged neutrophils represent an overly active subset exhibiting enhanced αMβ2 integrin activation and neutrophil extracellular trap formation under inflammatory conditions. Neutrophil ageing is driven by the microbiota via Toll-like receptor and myeloid differentiation factor 88-mediated signalling pathways. Depletion of the microbiota significantly reduces the number of circulating aged neutrophils and dramatically improves the pathogenesis and inflammation-related organ damage in models of sickle-cell disease or endotoxin-induced septic shock. These results identify a role for the microbiota in regulating a disease-promoting neutrophil subset.
Collapse
Affiliation(s)
- Dachuan Zhang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Grace Chen
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Deepa Manwani
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Arthur Mortha
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, New York 10029, USA.,The Immunology Institute, Mount Sinai School of Medicine, New York, New York 10029, USA
| | - Chunliang Xu
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Jeremiah J Faith
- The Immunology Institute, Mount Sinai School of Medicine, New York, New York 10029, USA.,The Institute for Genomics and Multiscale Biology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | - Robert D Burk
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Yuya Kunisaki
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Jung-Eun Jang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Christoph Scheiermann
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | - Miriam Merad
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, New York 10029, USA.,The Immunology Institute, Mount Sinai School of Medicine, New York, New York 10029, USA
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York 10461, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| |
Collapse
|
634
|
Jhunjhunwala S, Aresta-DaSilva S, Tang K, Alvarez D, Webber MJ, Tang BC, Lavin DM, Veiseh O, Doloff JC, Bose S, Vegas A, Ma M, Sahay G, Chiu A, Bader A, Langan E, Siebert S, Li J, Greiner DL, Newburger PE, von Andrian UH, Langer R, Anderson DG. Neutrophil Responses to Sterile Implant Materials. PLoS One 2015; 10:e0137550. [PMID: 26355958 PMCID: PMC4565661 DOI: 10.1371/journal.pone.0137550] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/18/2015] [Indexed: 12/20/2022] Open
Abstract
In vivo implantation of sterile materials and devices results in a foreign body immune response leading to fibrosis of implanted material. Neutrophils, one of the first immune cells to be recruited to implantation sites, have been suggested to contribute to the establishment of the inflammatory microenvironment that initiates the fibrotic response. However, the precise numbers and roles of neutrophils in response to implanted devices remains unclear. Using a mouse model of peritoneal microcapsule implantation, we show 30–500 fold increased neutrophil presence in the peritoneal exudates in response to implants. We demonstrate that these neutrophils secrete increased amounts of a variety of inflammatory cytokines and chemokines. Further, we observe that they participate in the foreign body response through the formation of neutrophil extracellular traps (NETs) on implant surfaces. Our results provide new insight into neutrophil function during a foreign body response to peritoneal implants which has implications for the development of biologically compatible medical devices.
Collapse
Affiliation(s)
- Siddharth Jhunjhunwala
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Stephanie Aresta-DaSilva
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Katherine Tang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - David Alvarez
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, 02115, United States of America
| | - Matthew J. Webber
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Benjamin C. Tang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Danya M. Lavin
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Omid Veiseh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Joshua C. Doloff
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Suman Bose
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Arturo Vegas
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Minglin Ma
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Gaurav Sahay
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Alan Chiu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Andrew Bader
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Erin Langan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Sean Siebert
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Jie Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Dale L. Greiner
- Department of Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts, 01605, United States of America
| | - Peter E. Newburger
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, 01605, United States of America
- Department of Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, 01605, United States of America
| | - Ulrich H. von Andrian
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, 02115, United States of America
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, 02139, United States of America
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
| | - Daniel G. Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States of America
- Department of Anesthesiology, Boston Children’s Hospital, Boston, Massachusetts, 02115, United States of America
- * E-mail:
| |
Collapse
|
635
|
|
636
|
Wound repair: role of immune-epithelial interactions. Mucosal Immunol 2015; 8:959-68. [PMID: 26174765 PMCID: PMC4916915 DOI: 10.1038/mi.2015.63] [Citation(s) in RCA: 208] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 05/28/2015] [Indexed: 02/07/2023]
Abstract
The epithelium serves as a highly selective barrier at mucosal surfaces. Upon injury, epithelial wound closure is orchestrated by a series of events that emanate from the epithelium itself as well as by the temporal recruitment of immune cells into the wound bed. Epithelial cells adjoining the wound flatten out, migrate, and proliferate to rapidly cover denuded surfaces and re-establish mucosal homeostasis. This process is highly regulated by proteins and lipids, proresolving mediators such as Annexin A1 protein and resolvins released into the epithelial milieu by the epithelium itself and infiltrating innate immune cells including neutrophils and macrophages. Failure to achieve these finely tuned processes is observed in chronic inflammatory diseases that are associated with non-healing wounds. An improved understanding of mechanisms that mediate repair is important in the development of therapeutics aimed to promote mucosal wound repair.
Collapse
|
637
|
Marques PE, Oliveira AG, Chang L, Paula-Neto HA, Menezes GB. Understanding liver immunology using intravital microscopy. J Hepatol 2015; 63:733-42. [PMID: 26055800 DOI: 10.1016/j.jhep.2015.05.027] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/21/2015] [Accepted: 05/21/2015] [Indexed: 12/15/2022]
Abstract
The liver has come a long way since it was considered only a metabolic organ attached to the gastrointestinal tract. The simultaneous ascension of immunology and intravital microscopy evidenced the liver as a central axis in the immune system, controlling immune responses to local and systemic agents as well as disease tolerance. The multiple hepatic cell populations are organized in a vascular environment that promotes intimate cellular interactions, including initiation of innate and adaptive immune responses, rapid leukocyte recruitment, pathogen clearance and production of a variety of immune mediators. In this review, we focus on the advances in liver immunology supported by intravital microscopy in diseases such as isquemia/reperfusion, acute liver injury and infections.
Collapse
Affiliation(s)
- Pedro Elias Marques
- Laboratório de Imunobiofotônica, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | - André Gustavo Oliveira
- Laboratório de Imunobiofotônica, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil
| | | | - Heitor Affonso Paula-Neto
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Brazil
| | - Gustavo Batista Menezes
- Laboratório de Imunobiofotônica, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
638
|
Seri Y, Shoda H, Suzuki A, Matsumoto I, Sumida T, Fujio K, Yamamoto K. Peptidylarginine deiminase type 4 deficiency reduced arthritis severity in a glucose-6-phosphate isomerase-induced arthritis model. Sci Rep 2015; 5:13041. [PMID: 26293116 PMCID: PMC4544002 DOI: 10.1038/srep13041] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 07/16/2015] [Indexed: 12/29/2022] Open
Abstract
Peptidyl arginine deiminase 4 (PAD4) is an enzyme that is involved in protein citrullination, and is a target for anti-citrullinated peptide antibodies (ACPAs) in rheumatoid arthritis (RA). Genetic polymorphisms in the PADI4 gene encoding PAD4 are associated with RA susceptibility. We herein analyzed the roles of PADI4 in inflammatory arthritis using a glucose-6-phosphate isomerase (GPI)-induced arthritis (GIA) model in Padi4 knockout (KO) mice. Arthritis severity, serum anti-GPI antibody titers, and IL-6 concentrations were significantly reduced in Padi4 KO mice. The frequency of Th17 cells was decreased in GPI-immunized Padi4 KO mice, whereas WT and Padi4-deficient naïve CD4+ T cells displayed the same efficiencies for Th17 cell differentiation in vitro. In addition, the numbers of myeloid lineage cells were reduced with the increased expression of pro-apoptotic genes in GPI-immunized Padi4 KO mice. Furthermore, the survival of Padi4-deficient neutrophils was impaired in vitro. Our results suggest that PADI4 exacerbates arthritis with diverse immunological modifications.
Collapse
Affiliation(s)
- Yu Seri
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Akari Suzuki
- Laboratory for Rheumatic Diseases, SNP Research Center, The Institute of Physical and Chemical Research (RIKEN), 1-7-22 Suehirocho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| | - Isao Matsumoto
- Division of Clinical Immunology, Major of Advanced Biomedical Applications, Graduate School of Comprehensive Human Sciences, Tsukuba University, Tsukuba, Ibaraki, 305-8575, Japan
| | - Takayuki Sumida
- Division of Clinical Immunology, Major of Advanced Biomedical Applications, Graduate School of Comprehensive Human Sciences, Tsukuba University, Tsukuba, Ibaraki, 305-8575, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Laboratory for Rheumatic Diseases, SNP Research Center, The Institute of Physical and Chemical Research (RIKEN), 1-7-22 Suehirocho, Tsurumi-ku, Yokohama City, Kanagawa 230-0045, Japan
| |
Collapse
|
639
|
Asadollahi F, Mehrzad J, Chaichi MJ, Taghavi Razavizadeh A. Photoimmunological properties of borage in bovine neutrophil in vitro model. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2015; 151:270-5. [PMID: 26334939 DOI: 10.1016/j.jphotobiol.2015.08.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/10/2015] [Accepted: 08/17/2015] [Indexed: 11/25/2022]
Abstract
Borage (Echium amoenum fisch) is one of the most commonly used medicinal plants, and has long been used as a traditional herbal medicine for many (non)infectious diseases in Iran. Study on photoredox and photoimmunology of borage is little. Natural immunomodulatory plants with minimal adverse/toxic effects could help boost animal health and, ultimately, public health. To determine the effect of borage on the functions of key circulating innate immune cells, effects of borage extract (BE) on bovine neutrophils (PMN) photoredox and phagocytosis events were evaluated using an in vitro model system. Blood PMN isolated from healthy high yielding dairy cows (n = 8/treatment) were pre-incubated with BE and the impact on phagocytosis-dependent-and-independent cellular chemiluminescence (CL), phagocytosis, killing of Staphylococcus aureus (S. aureus) and Escherichia coli (E. coli), fluorescence-based PMN H2O2 production and necrosis were assessed. Relative to control (no BE) PMN, treatment with BE significantly increased phagocytosis-dependent-and-independent PMN CL (>10-15% increase). While BE also led to increased PMN H2O2 production, necrosis was also surprisingly higher in these cells. Phagocytosis and killing of both E. coli and S. aureus by PMN treated with BE was substantially higher than that by control PMN. The increased photoimmunobiological events especially intracellular CL, intracellular H2O2 formation, and phagocytic capacity of BE-treated PMN support the potential immunotherapeutic implications of borage and its components for particularly immunocompromised animals and humans.
Collapse
Affiliation(s)
- Firouzeh Asadollahi
- Immunology Section, Department of Pathobiology, Faculty of Veterinary Medicine, and Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Jalil Mehrzad
- Immunology Section, Department of Pathobiology, Faculty of Veterinary Medicine, and Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| | | | - Alireza Taghavi Razavizadeh
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
640
|
Identification of Predictive Early Biomarkers for Sterile-SIRS after Cardiovascular Surgery. PLoS One 2015; 10:e0135527. [PMID: 26263001 PMCID: PMC4532358 DOI: 10.1371/journal.pone.0135527] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 07/22/2015] [Indexed: 12/21/2022] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is a common complication after cardiovascular surgery that in severe cases can lead to multiple organ dysfunction syndrome and even death. We therefore set out to identify reliable early biomarkers for SIRS in a prospective small patient study for timely intervention. 21 Patients scheduled for planned cardiovascular surgery were recruited in the study, monitored for signs of SIRS and blood samples were taken to investigate biomarkers at pre-assigned time points: day of admission, start of surgery, end of surgery, days 1, 2, 3, 5 and 8 post surgery. Stored plasma and cryopreserved blood samples were analyzed for cytokine expression (IL1β, IL2, IL6, IL8, IL10, TNFα, IFNγ), other pro-inflammatory markers (sCD163, sTREM-1, ESM-1) and response to endotoxin. Acute phase proteins CRP, PCT and pro-inflammatory cytokines IL6 and IL8 were significantly increased (p<0.001) at the end of surgery in all patients but could not distinguish between groups. Normalization of samples revealed significant increases in IL1β changes (p<0.05) and decreased responses to endotoxin (p<0.01) in the SIRS group at the end of surgery. Soluble TREM-1 plasma concentrations were significantly increased in patients with SIRS (p<0.01). This small scale patient study could show that common sepsis markers PCT, CRP, IL6 and TNFα had low predictive value for early diagnosis of SIRS after cardiovascular surgery. A combination of normalized IL1β plasma levels, responses to endotoxin and soluble TREM-1 plasma concentrations at the end of surgery are predictive markers of SIRS development in this small scale study and could act as an indicator for starting early therapeutic interventions.
Collapse
|
641
|
Yang ZW, Meng XX, Xu P. Central role of neutrophil in the pathogenesis of severe acute pancreatitis. J Cell Mol Med 2015; 19:2513-20. [PMID: 26249268 PMCID: PMC4627557 DOI: 10.1111/jcmm.12639] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/20/2015] [Indexed: 02/06/2023] Open
Abstract
Severe acute pancreatitis (SAP) is an acute abdominal disease with the strong systemic inflammatory response, and rapidly progresses from a local pancreatic damage into multiple organ dysfunction. For many decades, the contributions of neutrophils to the pathology of SAP were traditionally thought to be the chemokine and cytokine cascades that accompany inflammation. In this review, we focus mainly on those recently recognized aspects of neutrophils in SAP processes. First, emerging evidence suggests that therapeutic interventions targeting neutrophils significantly lower tissue damage and protect against the occurrence of pancreatitis. Second, trypsin activation promotes the initial neutrophils recruitment into local pancreas, and subsequently neutrophils infiltration in turn triggers trypsin production. Finally, neutrophils have the unique ability to release neutrophil extracellular traps even in the absence of pathogens.
Collapse
Affiliation(s)
- Zhi-Wen Yang
- Pharmacy Department, Songjiang Hospital Affiliated the First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Xiao Meng
- Digestive Department, Songjiang Hospital Affiliated the First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Xu
- Digestive Department, Songjiang Hospital Affiliated the First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
642
|
Huang X, Li J, Dorta-Estremera S, Di Domizio J, Anthony SM, Watowich SS, Popkin D, Liu Z, Brohawn P, Yao Y, Schluns KS, Lanier LL, Cao W. Neutrophils Regulate Humoral Autoimmunity by Restricting Interferon-γ Production via the Generation of Reactive Oxygen Species. Cell Rep 2015; 12:1120-32. [PMID: 26257170 DOI: 10.1016/j.celrep.2015.07.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 06/12/2015] [Accepted: 07/09/2015] [Indexed: 02/07/2023] Open
Abstract
Here, we examine the mechanism by which plasmacytoid dendritic cells (pDCs) and type I interferons promote humoral autoimmunity. In an amyloid-induced experimental autoimmune model, neutrophil depletion enhanced anti-nuclear antibody development, which correlated with heightened IFN-γ production by natural killer (NK) cells. IFN-α/β produced by pDCs activated NK cells via IL-15 induction. Neutrophils released reactive oxygen species (ROS), which negatively modulated the levels of IL-15, thereby inhibiting IFN-γ production. Mice deficient in NADPH oxidase 2 produced increased amounts of IFN-γ and developed augmented titers of autoantibodies. Both the pDC-IFN-α/β pathway and IFN-γ were indispensable in stimulating humoral autoimmunity. Male NZB/W F1 mice expressed higher levels of superoxide than their female lupus-prone siblings, and depletion of neutrophils resulted in spontaneous NK cell and autoimmune B cell activation. Our findings suggest a regulatory role for neutrophils in vivo and highlight the importance of an NK-IFN-γ axis downstream of the pDC-IFN-α/β pathway in systemic autoimmunity.
Collapse
Affiliation(s)
- Xinfang Huang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Rheumatology, Renji Hospital, Shanghai Institute of Rheumatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Jingjing Li
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Stephanie Dorta-Estremera
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Jeremy Di Domizio
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Scott M Anthony
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Daniel Popkin
- Department of Dermatology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Zheng Liu
- MedImmune, LLC, Gaithersburg, MD 20878, USA
| | | | - Yihong Yao
- MedImmune, LLC, Gaithersburg, MD 20878, USA
| | - Kimberly S Schluns
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology and Cancer Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Wei Cao
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
643
|
Szilagyi K, Gazendam RP, van Hamme JL, Tool ATJ, van Houdt M, Vos WAJW, Verkuijlen P, Janssen H, Belot A, Juillard L, Förster-Waldl E, Boztug K, Kraal G, de Winther MPJ, Kuijpers TW, van den Berg TK. Impaired microbial killing by neutrophils from patients with protein kinase C delta deficiency. J Allergy Clin Immunol 2015; 136:1404-7.e1-10. [PMID: 26233929 DOI: 10.1016/j.jaci.2015.06.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 06/03/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Katka Szilagyi
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - Roel P Gazendam
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | - John L van Hamme
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Anton T J Tool
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Michel van Houdt
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Wilhelm A J W Vos
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul Verkuijlen
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Hans Janssen
- Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alexandre Belot
- Hôpital Femme Mère Enfant, Hospices Civils de Lyon and Université de Lyon, Lyon, France
| | - Laurent Juillard
- CarMen U1060 Université de Lyon, Lyon, France; Service de Néphrologie, H. E. Herriot Hospices Civils de Lyon, Lyon, France
| | - Elisabeth Förster-Waldl
- Divison of Neonatology, Paediatric Intensive Care & Neuropaediatrics, Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Kaan Boztug
- Divison of Neonatology, Paediatric Intensive Care & Neuropaediatrics, Department of Paediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Georg Kraal
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Menno P J de Winther
- Department of Medical Biochemistry, Experimental Vascular Biology, Academic Medical Center, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Hematology, Immunology and Infectious Disease, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timo K van den Berg
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
644
|
Ekpenyong AE, Toepfner N, Chilvers ER, Guck J. Mechanotransduction in neutrophil activation and deactivation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015. [PMID: 26211453 DOI: 10.1016/j.bbamcr.2015.07.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Mechanotransduction refers to the processes through which cells sense mechanical stimuli by converting them to biochemical signals and, thus, eliciting specific cellular responses. Cells sense mechanical stimuli from their 3D environment, including the extracellular matrix, neighboring cells and other mechanical forces. Incidentally, the emerging concept of mechanical homeostasis,long term or chronic regulation of mechanical properties, seems to apply to neutrophils in a peculiar manner, owing to neutrophils' ability to dynamically switch between the activated/primed and deactivated/deprimed states. While neutrophil activation has been known for over a century, its deactivation is a relatively recent discovery. Even more intriguing is the reversibility of neutrophil activation and deactivation. We review and critically evaluate recent findings that suggest physiological roles for neutrophil activation and deactivation and discuss possible mechanisms by which mechanical stimuli can drive the oscillation of neutrophils between the activated and resting states. We highlight several molecules that have been identified in neutrophil mechanotransduction, including cell adhesion and transmembrane receptors, cytoskeletal and ion channel molecules. The physiological and pathophysiological implications of such mechanically induced signal transduction in neutrophils are highlighted as a basis for future work. This article is part of a Special Issue entitled: Mechanobiology.
Collapse
Affiliation(s)
- Andrew E Ekpenyong
- Department of Physics, Creighton University, Omaha, NE 68178, USA; Biotechnology Center, Technische Universität Dresden, Dresden, Germany
| | - Nicole Toepfner
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany; Klinik und Poliklinik für Kinder- und Jugendmedizin, Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Edwin R Chilvers
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke's and Papworth Hospitals, Cambridge CB2 0QQ, UK
| | - Jochen Guck
- Biotechnology Center, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
645
|
Trivedi RN, Agarwal P, Kumawat M, Pesingi PK, Gupta VK, Goswami TK, Mahawar M. Methionine sulfoxide reductase A (MsrA) contributes to Salmonella Typhimurium survival against oxidative attack of neutrophils. Immunobiology 2015. [PMID: 26224245 DOI: 10.1016/j.imbio.2015.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Salmonella Typhimurium (ST) must evade neutrophil assault for infection establishment in the host. Myeloperoxidase generated HOCl is the key antimicrobial agent produced by the neutrophils; and methionine (Met) residues are the primary targets of this oxidant. Oxidation of Mets leads to methionine sulfoxide (Met-SO) formation and consequently compromises the protein function(s). Methionine sulfoxide reductase A (MsrA) reductively repairs Met-SO to Mets. In this manner, MsrA maintains the function(s) of key proteins which are important for virulence of ST and enhance the survival of this bacterium under oxidative stress. We constructed msrA gene deletion strain (ΔmsrA). The primers located in the flanking regions to ΔmsrA gene amplified 850 and 300 bp amplicons in ST and ΔmsrA strains, respectively. The ΔmsrA strain grew normally in in vitro broth culture. However, ΔmsrA strain showed high susceptibility (p<0.001) to very low concentrations of HOCl which was restored (at least in part) by plasmid based complementation. ΔmsrA strain was hypersensitive (than ST) to the granules isolated from neutrophils. Further, the ΔmsrA strain was significantly (p<0.05) more susceptible to neutrophil mediated killing.
Collapse
Affiliation(s)
- Raj Narayan Trivedi
- The Immunology Section, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh 243 122, India
| | - Pranjali Agarwal
- Division of Biochemistry, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh 243 122, India
| | - Manoj Kumawat
- Division of Biochemistry, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh 243 122, India
| | - Pavan Kumar Pesingi
- Division of Veterinary Public Health, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh 243 122, India
| | | | - Tapas Kumar Goswami
- The Immunology Section, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh 243 122, India
| | - Manish Mahawar
- Division of Biochemistry, Indian Veterinary Research Institute, Izatnagar, Uttar Pradesh 243 122, India.
| |
Collapse
|
646
|
Hirai H, Yokota A, Tamura A, Sato A, Maekawa T. Non-steady-state hematopoiesis regulated by the C/EBPβ transcription factor. Cancer Sci 2015; 106:797-802. [PMID: 25940801 PMCID: PMC4520629 DOI: 10.1111/cas.12690] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 04/26/2015] [Accepted: 04/27/2015] [Indexed: 02/04/2023] Open
Abstract
Steady-state hematopoiesis responds to extracellular stimuli to meet changing demands and also to pathologically altered intracellular signaling. Granulocyte production increases following infection or in response to cytokine stimulation, and activation of the CCAAT/enhancer-binding protein β (C/EBPβ) transcription factor is required for such stress-induced granulopoiesis, whereas C/EBPα plays a critical role in maintaining steady-state granulopoiesis. Different roles of these C/EBP transcription factors in different modes of hematopoiesis are evolutionally conserved from zebrafish to humans. In addition to reactions against infections, C/EBPβ is responsible for cancer-driven myelopoiesis, which promotes cancer progression, at least in part, by abrogating the immune response in the cancer microenvironment. The BCR–ABL fusion protein activates emergency-specific pathway of granulopoiesis by upregulating C/EBPβ. This in turn causes chronic phase chronic myeloid leukemia, which is characterized by myeloid expansion. The C/EBPβ transcription factor also plays a role in other hematological malignancies of both myeloid and lymphoid lineage origin. Thus, elucidation of the upstream and downstream networks surrounding C/EBPβ will lead to the development of novel therapeutic strategies for diseases mediated by non-steady-state hematopoiesis.
Collapse
Affiliation(s)
- Hideyo Hirai
- Department of Transfusion Medicine and Cell Therapy, Kyoto University HospitalKyoto, Japan
- Correspondence Hideyo Hirai, Department of Transfusion Medicine and Cell Therapy, Kyoto University Hospital, 54 Kawahara-cho, Shogo-in, Sakyo-ku, Kyoto 606-8507, Japan., Tel: +81-75-751-3630; Fax: +81-75-751-4283;, E-mail:
| | - Asumi Yokota
- Department of Transfusion Medicine and Cell Therapy, Kyoto University HospitalKyoto, Japan
| | - Akihiro Tamura
- Department of Transfusion Medicine and Cell Therapy, Kyoto University HospitalKyoto, Japan
| | - Atsushi Sato
- Department of Transfusion Medicine and Cell Therapy, Kyoto University HospitalKyoto, Japan
| | - Taira Maekawa
- Department of Transfusion Medicine and Cell Therapy, Kyoto University HospitalKyoto, Japan
| |
Collapse
|
647
|
New insights into neutrophil and Leishmania infantum in vitro immune interactions. Comp Immunol Microbiol Infect Dis 2015; 40:19-29. [DOI: 10.1016/j.cimid.2015.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 02/24/2015] [Accepted: 03/20/2015] [Indexed: 01/09/2023]
|
648
|
Herwald H, Egesten A. Foodies of Innate Immunity. J Innate Immun 2015; 7:331-2. [PMID: 25997480 DOI: 10.1159/000430800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
649
|
Inhibition of the lymphocyte metabolic switch by the oxidative burst of human neutrophils. Clin Sci (Lond) 2015; 129:489-504. [PMID: 25951298 DOI: 10.1042/cs20140852] [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: 12/22/2014] [Accepted: 05/07/2015] [Indexed: 01/09/2023]
Abstract
Activation of the phagocytic NADPH oxidase-2 (NOX-2) in neutrophils is a critical process in the innate immune system and is associated with elevated local concentrations of superoxide, hydrogen peroxide (H2O2) and hypochlorous acid. Under pathological conditions, NOX-2 activity has been implicated in the development of autoimmunity, indicating a role in modulating lymphocyte effector function. Notably, T-cell clonal expansion and subsequent cytokine production requires a metabolic switch from mitochondrial respiration to aerobic glycolysis. Previous studies demonstrate that H2O2 generated from activated neutrophils suppresses lymphocyte activation but the mechanism is unknown. We hypothesized that activated neutrophils would prevent the metabolic switch and suppress the effector functions of T-cells through a H2O2-dependent mechanism. To test this, we developed a model co-culture system using freshly isolated neutrophils and lymphocytes from healthy human donors. Extracellular flux analysis was used to assess mitochondrial and glycolytic activity and FACS analysis to assess immune function. The neutrophil oxidative burst significantly inhibited the induction of lymphocyte aerobic glycolysis, caused inhibition of oxidative phosphorylation and suppressed lymphocyte activation through a H2O2-dependent mechanism. Hydrogen peroxide and a redox cycling agent, DMNQ, were used to confirm the impact of H2O2 on lymphocyte bioenergetics. In summary, we have shown that the lymphocyte metabolic switch from mitochondrial respiration to glycolysis is prevented by the oxidative burst of neutrophils. This direct inhibition of the metabolic switch is then a likely mechanism underlying the neutrophil-dependent suppression of T-cell effector function.
Collapse
|
650
|
Hwang TL, Aljuffali IA, Hung CF, Chen CH, Fang JY. The impact of cationic solid lipid nanoparticles on human neutrophil activation and formation of neutrophil extracellular traps (NETs). Chem Biol Interact 2015; 235:106-14. [PMID: 25920576 DOI: 10.1016/j.cbi.2015.04.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 03/30/2015] [Accepted: 04/09/2015] [Indexed: 12/13/2022]
Abstract
Cationic solid lipid nanoparticles (cSLNs) are extensively employed as the nanocarriers for drug/gene targeting to tumors and the brain. Investigation into the possible immune response of cSLNs is still lacking. The aim of this study was to evaluate the impact of cSLNs upon the activation of human polymorphonuclear neutrophil cells (PMNs). The cytotoxicity, pro-inflammatory mediators, Ca(2+) mobilization, mitogen-activated protein kinases (MAPKs), and neutrophil extracellular traps (NETs) as the indicators of PMN stimulation were examined in this work. The cSLNs presented a diameter of 195 nm with a zeta potential of 44 mV. The cSLNs could interact with the cell membrane to produce a direct membrane lysis and the subsequent cytotoxicity according to lactate dehydrogenase (LDH) elevation. The interaction of cSLNs with the membrane also triggered a Ca(2+) influx, followed by the induction of oxidative stress and degranulation. The cationic nanoparticles elevated the levels of superoxide anion and elastase by 24- and 9-fold, respectively. The PMN activation by cSLNs promoted the phosphorylation of p38 and Jun-N-terminal kinases (JNK) but not extracellular signal-regulated kinases (ERK). The imaging of scanning electron microscopy (SEM) and immunofluorescence demonstrated the production of NETs by cSLNs. This phenomenon was not significant for the neutral SLNs (nSLNs), although histones in NETs also increased after treatment of nSLNs. Our results suggest an important role of cSLNs in governing the activation of human neutrophils.
Collapse
Affiliation(s)
- Tsong-Long Hwang
- Cell Pharmacology Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Ibrahim A Aljuffali
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Chi-Feng Hung
- School of Medicine, Fu Jen Catholic University, Hsinchuang, New Taipei City, Taiwan, Taiwan
| | - Chun-Han Chen
- Division of General Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kweishan, Taoyuan, Taiwan; Research Center for Industry of Human Ecology, Chang Gung University of Science and Technology, Kweishan, Taoyuan, Taiwan.
| |
Collapse
|