1
|
Kim TC, Park HJ, Lee SW, Park YH, Van Kaer L, Hong S. Alpha-galactosylceramide pre-treatment attenuates clinical symptoms of LPS-induced acute neuroinflammation by converting pathogenic iNKT cells to anti-inflammatory iNKT10 cells in the brain. Inflamm Res 2024; 73:1511-1527. [PMID: 39028491 DOI: 10.1007/s00011-024-01915-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Invariant natural killer T (iNKT) cells play protective or pathogenic roles in a variety of immune and inflammatory diseases. However, whether iNKT cells contribute to the progression of acute neuroinflammation remains unclear. Thus, we addressed this question with a mouse model of lipopolysaccharide (LPS)-induced acute neuroinflammation. METHODS For induction of acute neuroinflammation, wild-type (WT) C57BL/6 (B6) mice were injected intraperitoneally (i.p.) with LPS for either three or five consecutive days, and then these mice were analyzed for brain-infiltrating leukocytes or mouse behaviors, respectively. To examine the role of iNKT cell activation in LPS-induced neuroinflammation, mice were injected i.p. with the iNKT cell agonist α-galactosylceramide (α-GalCer) seven days prior to LPS treatment. Immune cells infiltrated into the brain during LPS-induced neuroinflammation were determined by flow cytometry. In addition, LPS-induced clinical behavior symptoms such as depressive-like behavior and memory impairment in mice were evaluated by the open field and Y-maze tests, respectively. RESULTS We found that iNKT cell-deficient Jα18 mutant mice display delayed disease progression and decreased leukocyte infiltration into the brain compared with WT mice, indicating that iNKT cells contribute to the pathogenesis of LPS-induced neuroinflammation. Since it has been reported that pre-treatment with α-GalCer, an iNKT cell agonist, can convert iNKT cells towards anti-inflammatory phenotypes, we next explored whether pre-activation of iNKT cells with α-GalCer can regulate LPS-induced neuroinflammation. Strikingly, we found that α-GalCer pre-treatment significantly delays the onset of clinical symptoms, including depression-like behavior and memory impairment, while decreasing brain infiltration of pro-inflammatory natural killer cells and neutrophils, in this model of LPS-induced neuroinflammation. Such anti-inflammatory effects of α-GalCer pre-treatment closely correlated with iNKT cell polarization towards IL4- and IL10-producing phenotypes. Furthermore, α-GalCer pre-treatment restored the expression of suppressive markers on brain regulatory T cells during LPS-induced neuroinflammation. CONCLUSION Our findings provide strong evidence that α-GalCer-induced pre-activation of iNKT cells expands iNKT10 cells, mitigating depressive-like behaviors and brain infiltration of inflammatory immune cells induced by LPS-induced acute neuroinflammation. Thus, we suggest the prophylactic potential of iNKT cells and α-GalCer against acute neuroinflammation.
Collapse
Affiliation(s)
- Tae-Cheol Kim
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul, 05006, South Korea
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul, 05006, South Korea
| | - Sung Won Lee
- Department of Biomedical Laboratory Science, College of Health and Biomedical Services, Sangji University, Wonju, 26339, South Korea
| | - Yun Hoo Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul, 05006, South Korea
| | - Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul, 05006, South Korea.
| |
Collapse
|
2
|
Coleman M, Orvis A, Brokaw A, Furuta A, Sharma K, Quach P, Bhullar A, Sanghavi R, Nguyen S, Sweeney E, Seepersaud R, Armistead B, Adams Waldorf KM, Rajagopal L. GBS hyaluronidase mediates immune suppression in a TLR2/4- and IL-10-dependent manner during pregnancy-associated infection. mBio 2023; 14:e0204923. [PMID: 37747229 PMCID: PMC10653848 DOI: 10.1128/mbio.02049-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 09/26/2023] Open
Abstract
IMPORTANCE Bacteria such as GBS can cause infections during pregnancy leading to preterm births, stillbirths, and neonatal infections. The interaction between host and bacterial factors during infections in the placenta is not fully understood. GBS secretes a hyaluronidase enzyme that is thought to digest host hyaluronan into immunosuppressive disaccharides that dampen TLR2/4 signaling, leading to increased bacterial dissemination and adverse outcomes. In this study, we show that GBS HylB mediates immune suppression and promotes bacterial infection during pregnancy that requires TLR2, TLR4, and IL-10. Understanding the interaction between host and bacterial factors can inform future therapeutic strategies to mitigate GBS infections.
Collapse
Affiliation(s)
- Michelle Coleman
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Austyn Orvis
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Alyssa Brokaw
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Anna Furuta
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Kavita Sharma
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Phoenicia Quach
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Avneet Bhullar
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Rhea Sanghavi
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Shayla Nguyen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Erin Sweeney
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Ravin Seepersaud
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Blair Armistead
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Kristina M. Adams Waldorf
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Lakshmi Rajagopal
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
3
|
Li J, Wang P, Zhou T, Jiang W, Wu H, Zhang S, Deng L, Wang H. Neuroprotective effects of interleukin 10 in spinal cord injury. Front Mol Neurosci 2023; 16:1214294. [PMID: 37492521 PMCID: PMC10363608 DOI: 10.3389/fnmol.2023.1214294] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 06/26/2023] [Indexed: 07/27/2023] Open
Abstract
Spinal cord injury (SCI) starts with a mechanical and/or bio-chemical insult, followed by a secondary phase, leading progressively to severe collapse of the nerve tissue. Compared to the peripheral nervous system, injured spinal cord is characterized by weak axonal regeneration, which leaves most patients impaired or paralyzed throughout lifetime. Therefore, confining, alleviating, or reducing the expansion of secondary injuries and promoting functional connections between rostral and caudal regions of lesion are the main goals of SCI therapy. Interleukin 10 (IL-10), as a pivotal anti-inflammatory and immunomodulatory cytokine, exerts a wide spectrum of positive effects in the treatment of SCI. The mechanisms underlying therapeutic effects mainly include anti-oxidative stress, limiting excessive inflammation, anti-apoptosis, antinociceptive effects, etc. Furthermore, IL-10 displays synergistic effects when combined with cell transplantation or neurotrophic factor, enhancing treatment outcomes. This review lists pleiotropic mechanisms underlying IL-10-mediated neuroprotection after SCI, which may offer fresh perspectives for clinical translation.
Collapse
Affiliation(s)
- Juan Li
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Pei Wang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Ting Zhou
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Wenwen Jiang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Hang Wu
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Shengqi Zhang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| | - Lingxiao Deng
- Department of Neurological Surgery, Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Hongxing Wang
- Department of Rehabilitation Medicine, Zhongda Hospital Southeast University, Nanjing, China
| |
Collapse
|
4
|
Flerlage T, Crawford JC, Allen EK, Severns D, Tan S, Surman S, Ridout G, Novak T, Randolph A, West AN, Thomas PG. Single cell transcriptomics identifies distinct profiles in pediatric acute respiratory distress syndrome. Nat Commun 2023; 14:3870. [PMID: 37391405 PMCID: PMC10313703 DOI: 10.1038/s41467-023-39593-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 06/21/2023] [Indexed: 07/02/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS), termed pediatric ARDS (pARDS) in children, is a severe form of acute respiratory failure (ARF). Pathologic immune responses are implicated in pARDS pathogenesis. Here, we present a description of microbial sequencing and single cell gene expression in tracheal aspirates (TAs) obtained longitudinally from infants with ARF. We show reduced interferon stimulated gene (ISG) expression, altered mononuclear phagocyte (MNP) transcriptional programs, and progressive airway neutrophilia associated with unique transcriptional profiles in patients with moderate to severe pARDS compared to those with no or mild pARDS. We additionally show that an innate immune cell product, Folate Receptor 3 (FOLR3), is enriched in moderate or severe pARDS. Our findings demonstrate distinct inflammatory responses in pARDS that are dependent upon etiology and severity and specifically implicate reduced ISG expression, altered macrophage repair-associated transcriptional programs, and accumulation of aged neutrophils in the pathogenesis of moderate to severe pARDS caused by RSV.
Collapse
Affiliation(s)
- Tim Flerlage
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - E Kaitlynn Allen
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Danielle Severns
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Shaoyuan Tan
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sherri Surman
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Granger Ridout
- Hartwell Center for Biotechnology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Tanya Novak
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
| | - Adrienne Randolph
- Department of Anesthesiology, Critical Care, and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Anaesthesia, Harvard Medical School, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Alina N West
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
5
|
Kang J, Mateu-Borrás M, Monroe HL, Sen-Kilic E, Miller SJ, Dublin SR, Huckaby AB, Yang E, Pyles GM, Nunley MA, Chapman JA, Amin MS, Damron FH, Barbier M. Monoclonal antibodies against lipopolysaccharide protect against Pseudomonas aeruginosa challenge in mice. Front Cell Infect Microbiol 2023; 13:1191806. [PMID: 37424774 PMCID: PMC10326049 DOI: 10.3389/fcimb.2023.1191806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2023] Open
Abstract
Pseudomonas aeruginosa is a common cause of hospital-acquired infections, including central line-associated bloodstream infections and ventilator-associated pneumonia. Unfortunately, effective control of these infections can be difficult, in part due to the prevalence of multi-drug resistant strains of P. aeruginosa. There remains a need for novel therapeutic interventions against P. aeruginosa, and the use of monoclonal antibodies (mAb) is a promising alternative strategy to current standard of care treatments such as antibiotics. To develop mAbs against P. aeruginosa, we utilized ammonium metavanadate, which induces cell envelope stress responses and upregulates polysaccharide expression. Mice were immunized with P. aeruginosa grown with ammonium metavanadate and we developed two IgG2b mAbs, WVDC-0357 and WVDC-0496, directed against the O-antigen lipopolysaccharide of P. aeruginosa. Functional assays revealed that WVDC-0357 and WVDC-0496 directly reduced the viability of P. aeruginosa and mediated bacterial agglutination. In a lethal sepsis model of infection, prophylactic treatment of mice with WVDC-0357 and WVDC-0496 at doses as low as 15 mg/kg conferred 100% survival against challenge. In both sepsis and acute pneumonia models of infection, treatment with WVDC-0357 and WVDC-0496 significantly reduced bacterial burden and inflammatory cytokine production post-challenge. Furthermore, histopathological examination of the lungs revealed that WVDC-0357 and WVDC-0496 reduced inflammatory cell infiltration. Overall, our results indicate that mAbs directed against lipopolysaccharide are a promising therapy for the treatment and prevention of P. aeruginosa infections.
Collapse
Affiliation(s)
- Jason Kang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Margalida Mateu-Borrás
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Hunter L. Monroe
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University, Morgantown, WV, United States
| | - Emel Sen-Kilic
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Sarah Jo Miller
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Spencer R. Dublin
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Annalisa B. Huckaby
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Evita Yang
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Gage M. Pyles
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Mason A. Nunley
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Josh A. Chapman
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Md Shahrier Amin
- Department of Pathology, Anatomy, and Laboratory Medicine, West Virginia University, Morgantown, WV, United States
| | - F. Heath Damron
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Mariette Barbier
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Vaccine Development Center, West Virginia University Health Sciences Center, Morgantown, WV, United States
| |
Collapse
|
6
|
Paulsson M, Cardenas EI, Che KF, Brundin B, Smith M, Qvarfordt I, Lindén A. TLR4-mediated release of heparin-binding protein in human airways: a co-stimulatory role for IL-26. Front Immunol 2023; 14:1178135. [PMID: 37234157 PMCID: PMC10206387 DOI: 10.3389/fimmu.2023.1178135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Background Bacterial infection causes accumulation of neutrophils that release antimicrobial proteins including heparin-binding protein (HBP). In human airways, this neutrophil accumulation can be re-capitulated via intrabronchial exposure to lipopolysaccharide (LPS), a Toll-like receptor 4 (TLR4) agonist, that also causes a local increase in the neutrophil-mobilizing cytokine IL-26. Although LPS is considered a weak stimulus for HBP release ex vivo, its effect on HBP release in human airways in vivo has not been characterized. Methods We determined whether intrabronchial exposure to LPS causes concomitant release of HBP and IL-26 in human airways, and whether IL-26 can enhance LPS-induced release of HBP in isolated human neutrophils. Results We found that the concentration of HBP was markedly increased in bronchoalveolar lavage (BAL) fluid 12, 24, and 48 hours after LPS exposure, and that it displayed a strong and positive correlation with that of IL-26. Moreover, the concentration of HBP in conditioned media from isolated neutrophils was enhanced only after co-stimulation with LPS and IL-26. Conclusions Taken together, our findings indicate that TLR4 stimulation causes concomitant release of HBP and IL-26 in human airways, and that IL-26 may constitute a required co-stimulant for HBP release in neutrophils, thus enabling the concerted action of HBP and IL-26 in local host defense.
Collapse
Affiliation(s)
- Magnus Paulsson
- Division of Infection Medicine, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Clinical Microbiology, Laboratory Medicine, Skåne University Hospital, Lund, Sweden
| | - Eduardo I. Cardenas
- Division of Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karlhans F. Che
- Division of Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bettina Brundin
- Division of Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Margaretha Smith
- Division of Respiratory Medicine and Allergology, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingemar Qvarfordt
- Division of Respiratory Medicine and Allergology, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anders Lindén
- Division of Lung and Airway Research, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
7
|
Cartwright IM, Colgan SP. The hypoxic tissue microenvironment as a driver of mucosal inflammatory resolution. Front Immunol 2023; 14:1124774. [PMID: 36742292 PMCID: PMC9890178 DOI: 10.3389/fimmu.2023.1124774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
On the backdrop of all acute inflammatory processes lies the activation of the resolution response. Recent years have witnessed an emerging interest in defining molecular factors that influence the resolution of inflammation. A keystone feature of the mucosal inflammatory microenvironment is hypoxia. The gastrointestinal tract, particularly the colon, exists in a state of physiological hypoxia and during active inflammation, this hypoxic state is enhanced as a result of infiltrating leukocyte oxygen consumption and the activation of oxygen consuming enzymes. Most evidence suggests that mucosal hypoxia promotes the active resolution of inflammation through a variety of mechanisms, including extracellular acidification, purine biosynthesis/salvage, the generation of specialized pro-resolving lipid mediators (ie. resolvins) and altered chemokine/cytokine expression. It is now appreciated that infiltrating innate immune cells (neutrophils, eosinophils, macrophages) have an important role in molding the tissue microenvironment to program an active resolution response. Structural or functional dysregulation of this inflammatory microenvironment can result in the loss of tissue homeostasis and ultimately progression toward chronicity. In this review, we will discuss how inflammatory hypoxia drives mucosal inflammatory resolution and its impact on other microenvironmental factors that influence resolution.
Collapse
Affiliation(s)
- Ian M. Cartwright
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Sean P. Colgan
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, United States
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, CO, United States
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| |
Collapse
|
8
|
Carpinelli NA, Halfen J, Michelotti TC, Rosa F, Trevisi E, Chapman JD, Sharman ES, Osorio JS. Yeast Culture Supplementation Effects on Systemic and Polymorphonuclear Leukocytes' mRNA Biomarkers of Inflammation and Liver Function in Peripartal Dairy Cows. Animals (Basel) 2023; 13:ani13020301. [PMID: 36670844 PMCID: PMC9854537 DOI: 10.3390/ani13020301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
This study evaluated the effects of feeding a commercial yeast culture on blood biomarkers and polymorphonuclear leukocyte (PMNL) gene expression in dairy cows during the transition period until 50 d postpartum. Forty Holstein dairy cows were used in a randomized complete block design from -30 to 50 d. At -30 d, cows were assigned to a basal diet plus 114 g/d of top-dressed ground corn (control; n = 20) or 100 g/d of ground corn and 14 g/d of a yeast culture product (YC; n = 20). Blood samples were collected at various time points from -30 to 30 DIM to evaluate blood biomarkers and PMNL gene expression related to inflammation, liver function, and immune response. Liver function biomarkers, gamma-glutamyl transferase (GGT) and albumin were greater and lower, respectively, in YC cows in comparison to control. However, these biomarkers remained within physiological levels, indicating an active inflammatory process. Genes in PMNL expression related to inflammation (NFKB1, TNFA, TRAF6), anti-inflammation (IL10), and cell membrane receptors (SELL) were upregulated in the YC group in comparison to control. These results suggest that YC could stimulate a more active inflammatory response with signs of a resolution of inflammation in transition cows.
Collapse
Affiliation(s)
- Nathaly Ana Carpinelli
- Department of Dairy and Food Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Jessica Halfen
- Department of Dairy and Food Sciences, South Dakota State University, Brookings, SD 57007, USA
- Nucleo de Pesquisa, Ensino e Extensao em Pecuaria, Universidade Federal de Pelotas, Pelotas 96010610, Rio Grande do Sul, Brazil
| | | | - Fernanda Rosa
- Department of Dairy and Food Sciences, South Dakota State University, Brookings, SD 57007, USA
| | - Erminio Trevisi
- Department of Animal Sciences, Food and Nutrition (DIANA), Facoltà di Scienze Agrarie, Alimentari e Ambientali, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | | | | | - Johan S. Osorio
- Department of Dairy and Food Sciences, South Dakota State University, Brookings, SD 57007, USA
- Correspondence: ; Tel.: +1-5402311710
| |
Collapse
|
9
|
Montero‐Blay A, Blanco JD, Rodriguez‐Arce I, Lastrucci C, Piñero‐Lambea C, Lluch‐Senar M, Serrano L. Bacterial expression of a designed single-chain IL-10 prevents severe lung inflammation. Mol Syst Biol 2023; 19:e11037. [PMID: 36598022 PMCID: PMC9834763 DOI: 10.15252/msb.202211037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/27/2022] [Accepted: 10/05/2022] [Indexed: 01/05/2023] Open
Abstract
Interleukin-10 (IL-10) is an anti-inflammatory cytokine that is active as a swapped domain dimer and is used in bacterial therapy of gut inflammation. IL-10 can be used as treatment of a wide range of pulmonary diseases. Here we have developed a non-pathogenic chassis (CV8) of the human lung bacterium Mycoplasma pneumoniae (MPN) to treat lung diseases. We find that IL-10 expression by MPN has a limited impact on the lung inflammatory response in mice. To solve these issues, we rationally designed a single-chain IL-10 (SC-IL10) with or without surface mutations, using our protein design software (ModelX and FoldX). As compared to the IL-10 WT, the designed SC-IL10 molecules increase the effective expression in MPN four-fold, and the activity in mouse and human cell lines between 10 and 60 times, depending on the cell line. The SC-IL10 molecules expressed in the mouse lung by CV8 in vivo have a powerful anti-inflammatory effect on Pseudomonas aeruginosa lung infection. This rational design strategy could be used to other molecules with immunomodulatory properties used in bacterial therapy.
Collapse
Affiliation(s)
- Ariadna Montero‐Blay
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Javier Delgado Blanco
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Irene Rodriguez‐Arce
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Claire Lastrucci
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Carlos Piñero‐Lambea
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Maria Lluch‐Senar
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
| | - Luis Serrano
- Centre for Genomic Regulation (CRG)The Barcelona Institute of Science and TechnologyBarcelonaSpain
- Universitat Pompeu Fabra (UPF)BarcelonaSpain
- ICREABarcelonaSpain
| |
Collapse
|
10
|
Chokesuwattanaskul S, Fresneda Alarcon M, Mangalakumaran S, Grosman R, Cross AL, Chapman EA, Mason D, Moots RJ, Phelan MM, Wright HL. Metabolic Profiling of Rheumatoid Arthritis Neutrophils Reveals Altered Energy Metabolism That Is Not Affected by JAK Inhibition. Metabolites 2022; 12:650. [PMID: 35888774 PMCID: PMC9321732 DOI: 10.3390/metabo12070650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 02/05/2023] Open
Abstract
Neutrophils play a key role in the pathophysiology of rheumatoid arthritis (RA) where release of ROS and proteases directly causes damage to joints and tissues. Neutrophil function can be modulated by Janus Kinase (JAK) inhibitor drugs, including tofacitinib and baricitinib, which are clinically effective treatments for RA. However, clinical trials have reported increased infection rates and transient neutropenia during therapy. The subtle differences in the mode of action, efficacy and safety of JAK inhibitors have been the primary research topic of many clinical trials and systematic reviews, to provide a more precise and targeted treatment to patients. The aim of this study was to determine both the differences in the metabolome of neutrophils from healthy controls and people with RA, and the effect of different JAK inhibitors on the metabolome of healthy and RA neutrophils. Isolated neutrophils from healthy controls (HC) (n = 6) and people with RA (n = 7) were incubated with baricitinib, tofacitinib or a pan-JAK inhibitor (all 200 ng/mL) for 2 h. Metabolites were extracted, and 1H nuclear magnetic resonance (NMR) was applied to study the metabolic changes. Multivariate analyses and machine learning models showed a divergent metabolic pattern in RA neutrophils compared to HC at 0 h (F1 score = 86.7%) driven by energy metabolites (ATP, ADP, GTP and glucose). No difference was observed in the neutrophil metabolome when treated with JAK inhibitors. However, JAK inhibitors significantly inhibited ROS production and baricitinib decreased NET production (p < 0.05). Bacterial killing was not impaired by JAK inhibitors, indicating that the effect of JAK inhibitors on neutrophils can inhibit joint damage in RA without impairing host defence. This study highlights altered energy metabolism in RA neutrophils which may explain the cause of their dysregulation in inflammatory disease.
Collapse
Affiliation(s)
| | - Michele Fresneda Alarcon
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (M.F.A.); (A.L.C.); (E.A.C.)
| | | | - Rudi Grosman
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK; (R.G.); (M.M.P.)
- High Field NMR Facility, Liverpool Shared Research Facilities University of Liverpool, Liverpool L69 7TX, UK
| | - Andrew L. Cross
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (M.F.A.); (A.L.C.); (E.A.C.)
| | - Elinor A. Chapman
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (M.F.A.); (A.L.C.); (E.A.C.)
| | - David Mason
- Centre for Cell Imaging, Institute of Integrative Biology, University of Liverpool, Liverpool L69 7TX, UK;
| | - Robert J. Moots
- Department of Rheumatology, Aintree University Hospital, Liverpool L9 7AL, UK;
- Faculty of Health, Social Care and Medicine, Edge Hill University, Ormskirk L39 4QP, UK
| | - Marie M. Phelan
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK; (R.G.); (M.M.P.)
- High Field NMR Facility, Liverpool Shared Research Facilities University of Liverpool, Liverpool L69 7TX, UK
| | - Helen L. Wright
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; (M.F.A.); (A.L.C.); (E.A.C.)
| |
Collapse
|
11
|
Pretreatment with 6-Gingerol Ameliorates Sepsis-Induced Immune Dysfunction by Regulating the Cytokine Balance and Reducing Lymphocyte Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2021:5427153. [PMID: 35003518 PMCID: PMC8731291 DOI: 10.1155/2021/5427153] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/04/2021] [Accepted: 11/26/2021] [Indexed: 11/19/2022]
Abstract
Sepsis is characterized by an initial net hyperinflammatory response, followed by a period of immunosuppression, termed immunoparalysis. During this immunosuppressive phase, patients may have difficulty eradicating invading pathogens and are susceptible to life-threatening secondary hospital-acquired infections. Due to progress in antimicrobial treatment and supportive care, most patients survive early sepsis. Mortality is more frequently attributed to subsequent secondary nosocomial infections and multiorgan system failure. 6-Gingerol is the major pharmacologically active component of ginger. Although it is known to exhibit a variety of biological activities, including anti-inflammation and antioxidation, the role of 6-gingerol in sepsis-induced immune dysfunction remains elusive. Thus, we investigated whether 6-gingerol improves septic host response to infections during sepsis. 6-Gingerol-treated mice showed significantly lower mortality in polymicrobial sepsis induced by cecal ligation and puncture LPS via enhanced bacterial clearance in the peritoneum, blood, and organs (liver, spleen, and kidney) and inhibited the production of TNF-α and IL-6 in TLR2 and/or TLR4-stimulated macrophages. In addition, we demonstrated that survival improvement of secondary infection following septic insult was associated with an initial response of enhanced neutrophil numbers and function at the infection site, reduced apoptosis of immune cells, and a shift from a T helper cell type 2 (Th2) to a T helper cell type 1 (Th1) cytokine balance in the hypoinflammation phase. Our overall findings suggest that 6-gingerol potentially restores sepsis-induced immune dysfunction by shifting the balance of Th1/Th2 and by regulating apoptosis of immune cells.
Collapse
|
12
|
Moriarity DP, Ellman LM, Coe CL, Olino TM, Alloy LB. A physiometric investigation of inflammatory composites: Comparison of "a priori" aggregates, empirically-identified factors, and individual proteins. Brain Behav Immun Health 2021; 18:100391. [PMID: 34877552 PMCID: PMC8628205 DOI: 10.1016/j.bbih.2021.100391] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/15/2021] [Indexed: 01/03/2023] Open
Abstract
Most research testing the association between inflammation and health outcomes (e.g., heart disease, diabetes, depression) has focused on individual proteins; however, some studies have used summed composites of inflammatory markers without first investigating dimensionality. Using two different samples (MIDUS-2: N = 1255 adults, MIDUS-R: N = 863 adults), this study investigates the dimensionality of eight inflammatory proteins (C-reactive protein (CRP), interleukin (IL)-6, IL-8, IL-10, tumor necrosis factor-α (TNF-α), fibrinogen, E-selectin, and intercellular adhesion molecule (ICAM)-1) and compared the resulting factor structure to a) an "a priori"/tau-equivalent factor structure in which all inflammatory proteins equally load onto a single dimension (comparable to the summed composites) and b) proteins modeled individually (i.e., no latent variable) in terms of model fit, replicability, reliability, and their associations with health outcomes. An exploratory factor analysis indicated a two-factor structure (Factor 1: CRP and fibrinogen; Factor 2: IL-8 and IL-10) in MIDUS-2 and was replicated in MIDUS-R. Results did not clearly indicate whether the empirically-identified factor structure or the individual proteins modeled without a latent variable had superior model fit, but both strongly outperformed the "a priori"/tau-equivalent structure (which did not achieve acceptable model fit in any models). Modeling the empirically-identified factors and individual proteins (without a latent factor) as outcomes of medical diagnoses resulted in comparable conclusions. However, modeling individual proteins resulted in findings more robust to correction for multiple comparisons despite more conservative adjustments. Further, reliability for all latent variables was poor. These results indicate that modeling inflammation as a unidimensional construct equally associated with all available proteins does not fit the data well. Instead, individual inflammatory proteins or, potentially (if empirically supported and biologically-plausible) empirically-identified inflammatory factors should be used in accordance with theory.
Collapse
Affiliation(s)
- Daniel P. Moriarity
- Temple University, USA
- McLean Hospital, Harvard Medical School
- Corresponding author. Department of Psychology, Temple University, Weiss Hall, 1701 N. 13th St., Philadelphia, PA 19122, USA.
| | | | | | | | | |
Collapse
|
13
|
Wijers CDM, Pham L, Menon S, Boyd KL, Noel HR, Skaar EP, Gaddy JA, Palmer LD, Noto MJ. Identification of Two Variants of Acinetobacter baumannii Strain ATCC 17978 with Distinct Genotypes and Phenotypes. Infect Immun 2021; 89:e0045421. [PMID: 34460288 PMCID: PMC8594612 DOI: 10.1128/iai.00454-21] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 01/11/2023] Open
Abstract
Acinetobacter baumannii is a nosocomial pathogen that exhibits substantial genomic plasticity. Here, the identification of two variants of A. baumannii ATCC 17978 that differ based on the presence of a 44-kb accessory locus, named AbaAL44 (A. baumannii accessory locus 44 kb), is described. Analyses of existing deposited data suggest that both variants are found in published studies of A. baumannii ATCC 17978 and that American Type Culture Collection (ATCC)-derived laboratory stocks comprise a mix of these two variants. Yet, each variant exhibits distinct interactions with the host in vitro and in vivo. Infection with the variant that harbors AbaAL44 (A. baumannii 17978 UN) results in decreased bacterial burdens and increased neutrophilic lung inflammation in a mouse model of pneumonia, and affects the production of interleukin 1 beta (IL-1β) and IL-10 by infected macrophages. AbaAL44 harbors putative pathogenesis genes, including those predicted to encode a type I pilus cluster, a catalase, and a cardiolipin synthase. The accessory catalase increases A. baumannii resistance to oxidative stress and neutrophil-mediated killing in vitro. The accessory cardiolipin synthase plays a dichotomous role by promoting bacterial uptake and increasing IL-1β production by macrophages, but also by enhancing bacterial resistance to cell envelope stress. Collectively, these findings highlight the phenotypic consequences of the genomic dynamism of A. baumannii through the evolution of two variants of a common type strain with distinct infection-related attributes.
Collapse
Affiliation(s)
- Christiaan D. M. Wijers
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ly Pham
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Swapna Menon
- AnalyzeDat Consulting Services, Ernakulam, Kerala, India
| | - Kelli L. Boyd
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hannah R. Noel
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jennifer A. Gaddy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee, USA
| | - Lauren D. Palmer
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Michael J. Noto
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
14
|
Weathered C, Pennington K, Escalante P, Pienaar E. The Role of Biofilms, Bacterial Phenotypes, and Innate Immune Response in Mycobacterium avium Colonization to Infection. J Theor Biol 2021; 534:110949. [PMID: 34717938 DOI: 10.1016/j.jtbi.2021.110949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 01/15/2023]
Abstract
Mycobacterium avium complex (MAC), is known for colonizing and infecting humans following inhalation of the bacteria. MAC pulmonary disease is notoriously difficult to treat and prone to recurrence. Both the incidence and prevalence MAC pulmonary disease have been increasing globally. MAC is well known to form biofilms in the environment, and in vitro, these biofilms have been shown to aid MAC in epithelial cell invasion, protect MAC from phagocytosis, and cause premature apoptosis in macrophages. In vivo, the system of interactions between MAC, biofilms and host macrophages is complex, difficult to replicate in vitro and in animal models, has not been fully characterized. Here we present a three-dimensional agent-based model of a lung airway to help understand how these interactions evolve in the first 14 days post-bacterial inhalation. We parameterized the model using published data and performed uncertainty analysis to characterize outcomes and parameters' effects on those outcomes. Model results show diverse outcomes, including wide ranges of macrophage recruitment levels, and bacterial loads and phenotype distribution. Though most bacteria are phagocytosed by macrophages and remain intracellular, there are also many simulations in which extracellular bacteria continue to drive the colonization and infection. Initial parameters dictating host immune levels, bacterial loads introduced to the airway, and biofilm conditions have significant and lasting impacts on the course of these results. Additionally, though macrophage recruitment is key for suppressing bacterial loads, there is evidence of significant excess recruitment that fail to impact bacterial numbers. These results highlight a need and identify a path for further exploration into the inhalation events in MAC infection. Early infection dynamics could have lasting impacts on the development of nodular bronchiectatic or fibrocavitary disease as well as inform possible preventative and treatment intervention targeting biofilm-macrophage interactions.
Collapse
Affiliation(s)
- Catherine Weathered
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| | - Kelly Pennington
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Patricio Escalante
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Elsje Pienaar
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| |
Collapse
|
15
|
Deng L, Zhao M, Wang Y, Wang X, Liu J. Dexmedetomidine Inhibits Acute Lung Injury by Upregulating miR-144 Expression in Mice. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The understanding of lung injury’s mechanisms at the molecular level is not fully completed. MicroR-NAs (miRNAs), which are part of different pathophysiological processes, are essential biological regulators that operate by suppressing target genes. A mouse model of acute lung
injury (ALI), which is triggered by lipopolysaccharide (LPS), was used to analyze miR-144 level in the ALI mice with or without dexmedetomidine treatment. Inflammation was investigated by the ratio of wet weight’s value to dry weight (W/D) of the lung, the release of cytokines TNF-α,
cytokines IL-6, and cytokines IL-1β, and MPO activity. To validate the effect of dexmedetomidine on miR-144, overex-pression and knockdown of miR-144 were applied to treat antagomir144 and agomir144. The result suggested that LPS-triggered ALI was alleviated by dexmedetomidine.
miR-144 was downregulated in ALI mice. The knockdown of miR-144 attenuated the protection of dexmedetomidine to acute lung injury. Overexpression of miR-144 attenuated the ALI, which was induced by LPS.
Collapse
Affiliation(s)
- Liqiang Deng
- Shandong Provincial Maternal and Child Health Care Hospital, Jinan 250014, China
| | - Min Zhao
- Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao 266000, China
| | - Yihao Wang
- Qingdao Municipal Hospital, Qingdao 266000, China
| | - Xujian Wang
- Shandong Provincial Maternal and Child Health Care Hospital, Jinan 250014, China
| | - Juan Liu
- Shandong Provincial Maternal and Child Health Care Hospital, Jinan 250014, China
| |
Collapse
|
16
|
Qin W, Brands X, van’t Veer C, F. de Vos A, Sirard JC, J. T. H. Roelofs J, P. Scicluna B, van der Poll T. Bronchial epithelial DNA methyltransferase 3b dampens pulmonary immune responses during Pseudomonas aeruginosa infection. PLoS Pathog 2021; 17:e1009491. [PMID: 33793661 PMCID: PMC8043394 DOI: 10.1371/journal.ppat.1009491] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 04/13/2021] [Accepted: 03/22/2021] [Indexed: 01/01/2023] Open
Abstract
DNA methyltransferase (Dnmt)3b mediates de novo DNA methylation and modulation of Dnmt3b in respiratory epithelial cells has been shown to affect the expression of multiple genes. Respiratory epithelial cells provide a first line of defense against pulmonary pathogens and play a crucial role in the immune response during pneumonia caused by Pseudomonas (P.) aeruginosa, a gram-negative bacterium that expresses flagellin as an important virulence factor. We here sought to determine the role of Dntm3b in respiratory epithelial cells in immune responses elicited by P. aeruginosa. DNMT3B expression was reduced in human bronchial epithelial (BEAS-2B) cells as well as in primary human and mouse bronchial epithelial cells grown in air liquid interface upon exposure to P. aeruginosa (PAK). Dnmt3b deficient human bronchial epithelial (BEAS-2B) cells produced more CXCL1, CXCL8 and CCL20 than control cells when stimulated with PAK, flagellin-deficient PAK (PAKflic) or flagellin. Dnmt3b deficiency reduced DNA methylation at exon 1 of CXCL1 and enhanced NF-ĸB p65 binding to the CXCL1 promoter. Mice with bronchial epithelial Dntm3b deficiency showed increased Cxcl1 mRNA expression in bronchial epithelium and CXCL1 protein release in the airways during pneumonia caused by PAK, which was associated with enhanced neutrophil recruitment and accelerated bacterial clearance; bronchial epithelial Dnmt3b deficiency did not modify responses during pneumonia caused by PAKflic or Klebsiella pneumoniae (an un-flagellated gram-negative bacterium). Dnmt3b deficiency in type II alveolar epithelial cells did not affect mouse pulmonary defense against PAK infection. These results suggest that bronchial epithelial Dnmt3b impairs host defense during Pseudomonas induced pneumonia, at least in part, by dampening mucosal responses to flagellin.
Collapse
Affiliation(s)
- Wanhai Qin
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Xanthe Brands
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Cornelis van’t Veer
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Alex F. de Vos
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Claude Sirard
- Université de Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Joris J. T. H. Roelofs
- Department of Pathology, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Brendon P. Scicluna
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Tom van der Poll
- Center of Experimental & Molecular Medicine, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centers, location Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
17
|
Skeletal muscle fibers play a functional role in host defense during sepsis in mice. Sci Rep 2021; 11:7316. [PMID: 33795743 PMCID: PMC8016959 DOI: 10.1038/s41598-021-86585-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/11/2021] [Indexed: 12/19/2022] Open
Abstract
Skeletal muscles secrete a wide variety of immunologically active cytokines, but the functional significance of this response to in vivo innate immunity is not understood. We addressed this by knocking out the toll receptor adapter protein, Myd88, only in skeletal muscle fibers (skmMyd88KO), and followed male and female mice at 6 and 12 h after peritoneal injection of cecal slurry (CS), a model of polymicrobial sepsis. Because of a previously identified increase in mortality to CS injection, males received ~ 30% lower dose. At 12 h, skmMyd88KO caused significant reductions in a wide variety of pro- and anti-inflammatory plasma cytokines, e.g. TNFα, IL-1β and IL-10, compared to strain-matched controls in both males and females. Similar reductions were observed at 6 h in females. SkmMyd88KO led to ~ 40–50% elevations in peritoneal neutrophils at 6 and 12 h post CS in females. At 12 h post CS, skmMyd88KO increased peritoneal monocytes/macrophages and decreased %eosinophils and %basophils in females. SkmMyd88KO also led to significantly higher rates of mortality in female mice but not in males. In conclusion, the results suggest that skeletal muscle Myd88-dependent signal transduction can play functionally important role in normal whole body, innate immune inflammatory responses to peritoneal sepsis.
Collapse
|
18
|
Lewis BW, Choudhary I, Paudel K, Mao Y, Sharma R, Wang Y, Deshane JS, Boucher RC, Patial S, Saini Y. The Innate Lymphoid System Is a Critical Player in the Manifestation of Mucoinflammatory Airway Disease in Mice. THE JOURNAL OF IMMUNOLOGY 2020; 205:1695-1708. [PMID: 32817334 DOI: 10.4049/jimmunol.2000530] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/21/2020] [Indexed: 11/19/2022]
Abstract
Innate lymphoid and adaptive immune cells are known to regulate epithelial responses, including mucous cell metaplasia (MCM), but their roles in mucoinflammatory airway diseases, such as cystic fibrosis, remain unknown. Scnn1b transgenic (Scnn1b-Tg+) mice, which recapitulate cystic fibrosis-like mucoinflammatory airway disease, deficient in innate lymphoid (Il2rg knockout mice [Il2rg KO]), adaptive immune (Rag1 knockout mice [Rag1 KO]), or both systems (Il2rg KO/Rag1 KO), were employed to investigate their respective contributions in the pathogenesis of mucoinflammatory airway disease. As previously reported, immunocompetent Tg+ juveniles exhibited spontaneous neonatal bacterial infections with robust mucoinflammatory features, including elevated expression of Th2-associated markers accompanied by MCM, elevated MUC5B expression, and airway mucus obstruction. The bacterial burden was increased in Il2rg KO/Tg+ juveniles but returned to significantly lower levels in Il2rg KO/Rag1 KO/Tg+ juveniles. Mechanistically, this improvement reflected reduced production of adaptive immunity-derived IL-10 and, in turn, increased activation of macrophages. Although all the mucoinflammatory features were comparable between the immunocompetent Tg+ and Rag1 KO/Tg+ juveniles, the Il2rg KO/Tg+ and Il2rg KO/Rag1 KO/Tg+ juveniles exhibited suppressed expression levels of Th2 markers, diminished MCM, suppressed MUC5B expression, and reduced mucus obstruction. Collectively, these data indicate that, in the context of airway mucus obstruction, the adaptive immune system suppresses antibacterial macrophage activation, whereas the innate lymphoid system contributes to MCM, mucin production, and mucus obstruction.
Collapse
Affiliation(s)
- Brandon W Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Ishita Choudhary
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Kshitiz Paudel
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Yun Mao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Rahul Sharma
- National Hansen's Disease Program, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Yong Wang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Jessy S Deshane
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294; and
| | - Richard C Boucher
- Marsico Lung Institute/University of North Carolina Cystic Fibrosis Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803;
| |
Collapse
|
19
|
Hemorrhage Attenuates Neutrophil Recruitment in Response to Secondary Respiratory Infection by Pseudomonas Aeruginosa. Shock 2020; 52:506-512. [PMID: 30475329 DOI: 10.1097/shk.0000000000001288] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Neutrophil recruitment into the lung airspaces plays an important role in the containment and clearance of bacteria. Hemorrhagic shock, a complication of traumatic injury, induces immune dysfunction that compromises host defense and frequently leads to secondary infection. The objective of the current study was to determine whether prior hemorrhage impacts neutrophil recruitment in response to secondary Pseudomonas aeruginosa. Experiments were performed using a mouse model (C57BL/6) of respiratory infection by P. aeruginosa (strain PA103, 3 × 10 colony-forming units [CFUs]) that is delivered by intratracheal inhalation 24 h after hypovolemic hemorrhagic shock (fixed mean arterial blood pressure at 35 mmHg for 90 min, Ringer's lactate infused as fluid resuscitation). By postmortem flow cytometry analyses of bronchoalveolar lavage fluid, we observe that prior hemorrhage attenuates the entry of neutrophils into the lung airspaces in response to P. aeruginosa. The reduction in neutrophil recruitment occurs in an amplified inflammatory environment, with elevated lung tissue levels of interleukin 6 and C-X-C motif ligand 1 in mice receiving hemorrhage prior to infection. As compared to either insult alone, outcome to sequential hemorrhage and respiratory infection includes enhanced mortality. The effect of prior hemorrhage on clearance of P. aeruginosa, as determined by quantifying bacterial CFUs in lung tissue, was not statistically significant at 24 h postinfection, but our data suggest that further inquiry may be needed to fully understand the potential impact of hemorrhagic shock on this process. These results suggest that changes in neutrophil recruitment may contribute to the immune dysfunction following hemorrhagic shock that renders the host susceptible to severe respiratory infection.
Collapse
|
20
|
Ye C, Li H, Bao M, Zhuo R, Jiang G, Wang W. Alveolar macrophage - derived exosomes modulate severity and outcome of acute lung injury. Aging (Albany NY) 2020; 12:6120-6128. [PMID: 32259794 PMCID: PMC7185135 DOI: 10.18632/aging.103010] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/20/2020] [Indexed: 12/31/2022]
Abstract
Severe acute lung injury (ALI) can cause death, and the survivals may develop acute respiratory distress syndrome (ARDS) due to fibrotic repair of the lung. Alveolar macrophages play a demonstrative role during the pathogenesis of ALI, and the timing and degree of differentially polarization of macrophages determine the severity of disease and outcome. Exosomes are important mediators of cellular communication and play critical roles during macrophage differentiation, proliferation and function. Nevertheless, the exact effects of alveolar macrophage - derived exosomes on ALI remain unknow. Here, we used lipopolysaccharide (LPS) to induce ALI in mice and analyzed the exosome population in bronchoalveolar lavage fluid (BALF) from macrophages, neutrophils and epithelial cells at different time points after treatment. Our data showed that macrophages were the major secretors for early secreted pro-inflammatory cytokines in the BALF-exosomes, which likely activated neutrophils to produce a variety of pro-inflammatory cytokines and IL-10. IL-10 by neutrophils in BALF-exosomes likely in turn polarized macrophages to M2c, which may be responsible for post-ALI fibrosis. Our study thus reveals a previous non-acknowledged role of BALF-exosomes as a mediator of inflammatory response and cell crosstalk during ALI.
Collapse
Affiliation(s)
- Cong Ye
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Huiting Li
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Minwei Bao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Ran Zhuo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, Shanghai 200433, China
| | - Weixi Wang
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
21
|
Kang MJ, Jang AR, Park JY, Ahn JH, Lee TS, Kim DY, Lee MS, Hwang S, Jeong YJ, Park JH. IL-10 Protects Mice From the Lung Infection of Acinetobacter baumannii and Contributes to Bacterial Clearance by Regulating STAT3-Mediated MARCO Expression in Macrophages. Front Immunol 2020; 11:270. [PMID: 32153580 PMCID: PMC7047127 DOI: 10.3389/fimmu.2020.00270] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/03/2020] [Indexed: 11/13/2022] Open
Abstract
Interleukin-10 plays important, yet contrasting, roles in host protection against bacterial infections and in the septic response. To determine the role of IL-10 in the host defense against Acinetobacter baumannii infection, wild-type (WT) and IL-10-deficient mice were infected intranasally with the bacteria. IL-10-deficient mice exhibited increased mortality, severe pathology, and excess production of proinflammatory cytokines and chemokines in the lungs, and increased bacterial burdens in bronchoalveolar lavage (BAL) fluids and lung homogenates after A. baumannii infection, compared to WT mice. Intranasal administration of recombinant IL-10 rescued mice from the lethality of the bacterial infection by promoting bacterial clearance and reducing production of cytokines and chemokines in the lungs. In vitro experiments revealed that IL-10 enhanced phagocytosis and bacterial killing by macrophages by upregulating the macrophage receptor with collagenous structure (MARCO). In addition, A. baumannii-induced activation of STAT3 was impaired in IL-10-deficient macrophages, which was essential for expression of MARCO. Intranasal adoptive transfer of WT macrophages resulted in significant increases in mice survival and bacterial clearance in IL-10-deficient mice infected with A. baumannii. Our results show that IL-10 played an important role in the host defense against pulmonary infection of A. baumannii by promoting the antibacterial function of macrophages by regulating MARCO expression through the STAT3-mediated pathway.
Collapse
Affiliation(s)
- Min-Jung Kang
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Ah-Ra Jang
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Ji-Yeon Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Jae-Hun Ahn
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Tae-Sung Lee
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Dong-Yeon Kim
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Moo-Seung Lee
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Seungwoo Hwang
- Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Yu-Jin Jeong
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Jong-Hwan Park
- Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| |
Collapse
|
22
|
Arendt M, Elissa J, Schmidt N, Michael E, Potter N, Cook M, Knoll LJ. Investigating the role of interleukin 10 on Eimeria intestinal pathogenesis in broiler chickens. Vet Immunol Immunopathol 2019; 218:109934. [PMID: 31520870 DOI: 10.1016/j.vetimm.2019.109934] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 07/22/2019] [Accepted: 08/27/2019] [Indexed: 12/27/2022]
Abstract
Eimeria species are intestinal protozoan parasites that cause lack of production, malabsorption and mortality in floor raised chickens. Administering an oral antibody to interleukin 10 (aIL-10) reduces the symptoms of coccidiosis in broilers, indicating interleukin 10 (IL-10) is key to Eimeria pathology. IL-10 is an anti-inflammatory cytokine and acts as a stand down signal to reduce inflammation and host pathology during disease. Related protozoan parasites exploit IL-10 to reduce pathogen-damaging host inflammatory responses. We hypothesize that IL-10 is increased during Eimeria infection through an unknown host-pathogen interaction, and by feeding aIL-10 to neutralize excess IL-10 the bird is allowed to mount an effective immune response to Eimeria. To determine the effects of aIL-10 during the intestinal immune response, intestinal pathology and the relationship between IL-10, interferon gamma (IFNγ) and Eimeria infection were evaluated in this study. In both experiments, broilers were administered either a 10x dose of Advent® Eimeria vaccine or saline. Duodenum, jejunum and cecum samples were collected, processed, stained and examined under a microscope. Evaluation of intestinal histomorphology during aIL-10 administration showed minimal differences in birds fed aIL-10 during infection compared to animals fed a control antibody during Eimeria infection. To further evaluate aIL-10's positive effect during infection, immunofluorescent histochemistry was performed on chicken intestines days 3-7 post Eimeria infection for IL-10 and IFNγ presence in intestinal mucosa in control and infected birds, in regions with and without visible Eimeria burden. IL-10 and IFNγ had significant changes between days 4.5-7 post-infection in birds fed aIL-10 compared to animals fed a control antibody. Overall we found that the duodenum had increased IL-10 presence and increased IFNγ presence, and the jejunum and cecum had decreased IL-10 presence and decreased IFNγ presence. These differences in spatial regulation of IL-10 and IFNγ may indicate Eimeria species induce slightly different cytokine responses.
Collapse
Affiliation(s)
- Maria Arendt
- University of Wisconsin - Madison, Comparative Biomedical Sciences Department, United States.
| | - Jonathan Elissa
- University of Wisconsin - Madison, School of Veterinary Medicine, United States
| | - Natalie Schmidt
- University of Wisconsin - Madison, Animal Science Department, United States
| | - Emily Michael
- University of Wisconsin - Madison, Animal Science Department, United States
| | - Nicole Potter
- University of Wisconsin - Madison, Animal Science Department, United States
| | - Mark Cook
- University of Wisconsin - Madison, Animal Science Department, United States
| | - Laura J Knoll
- University of Wisconsin - Madison, Medical Microbiology & Immunology Department, United States
| |
Collapse
|
23
|
Hashemi FB, Behrouz B, Irajian G, Laghaei P, Korpi F, Fatemi MJ. A trivalent vaccine consisting of "flagellin A+B and pilin" protects against Pseudomonas aeruginosa infection in a murine burn model. Microb Pathog 2019; 138:103697. [PMID: 31465785 DOI: 10.1016/j.micpath.2019.103697] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 02/22/2019] [Accepted: 08/26/2019] [Indexed: 10/26/2022]
Abstract
Pseudomonas aeruginosa is a common nosocomial pathogen in burn patients, and rapidly achieves antibiotic resistance, and thus, developing an effective vaccine is critically important for combating P. aeruginosa infection. Flagella and pili play important roles in colonization of P. aeruginosa at the burn wound site and its subsequent dissemination to deeper tissue and organs. In the present study, we evaluated protective efficacy of a trivalent vaccine containing flagellins A and B (FlaA + FlaB) + pilin (PilA) in a murine burn model of infection. "FlaA + FlaB + PilA" induced greater protection in P. aeruginosa murine burn model than the single components alone, and it showed broad immune protection against P. aeruginosa strains. Immunization with "FlaA + FlaB + PilA" induced strong opsonophagocytic antibodies and resulted in reduced bacterial loads, systemic IL-12/IL-10 cytokine expression, and increased survival after challenge with three times lethal dose fifty (LD50) of P. eruginosa strains. Moreover, the protective efficacy of "FlaA + FlaB + PilA" vaccination was largely attributed to specific antibodies. Taken together, these data further confirm that the protective effects of "FlaA + FlaB + PilA" vaccine significantly enhance efficacy compared with antibodies against either mono or divalent antigen, and that the former broadens the coverage against P. eruginosa strains that express two of the three antigens.
Collapse
Affiliation(s)
- Farhad B Hashemi
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahador Behrouz
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | - Gholamreza Irajian
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parisa Laghaei
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Korpi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Fatemi
- Burn Research Center, Hazrat Fatima Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Valiate BVS, Alvarez RU, Karra L, Queiroz‐Júnior CM, Amaral FA, Levi‐Schaffer F, Teixeira MM. The immunoreceptor CD300a controls the intensity of inflammation and dysfunction in a model of Ag‐induced arthritis in mice. J Leukoc Biol 2019; 106:957-966. [DOI: 10.1002/jlb.3a1018-389r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 03/21/2019] [Accepted: 05/10/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
- Bruno V. S. Valiate
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Rodrigo U. Alvarez
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Laila Karra
- Pharmacology and Experimental Therapeutics UnitInstitute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem Israel
| | | | - Flavio A. Amaral
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Francesca Levi‐Schaffer
- Pharmacology and Experimental Therapeutics UnitInstitute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem Jerusalem Israel
| | - Mauro M. Teixeira
- Departamento de Bioquímica e ImunologiaInstituto de Ciências Biológicas, Universidade Federal de Minas Gerais Belo Horizonte Brazil
| |
Collapse
|
25
|
Mould KJ, Janssen WJ. Recombinant IFN-β for Postseptic Acute Lung Injury-What's the Mechanism? Am J Respir Cell Mol Biol 2019; 59:1-2. [PMID: 29957050 DOI: 10.1165/rcmb.2018-0054ed] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Kara J Mould
- 1 Department of Medicine National Jewish Health Denver, Colorado and.,2 Department of Medicine University of Colorado Denver Denver, Colorado
| | - William J Janssen
- 1 Department of Medicine National Jewish Health Denver, Colorado and.,2 Department of Medicine University of Colorado Denver Denver, Colorado
| |
Collapse
|
26
|
Sécher T, Dalonneau E, Ferreira M, Parent C, Azzopardi N, Paintaud G, Si-Tahar M, Heuzé-Vourc'h N. In a murine model of acute lung infection, airway administration of a therapeutic antibody confers greater protection than parenteral administration. J Control Release 2019; 303:24-33. [PMID: 30981816 DOI: 10.1016/j.jconrel.2019.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/06/2019] [Accepted: 04/03/2019] [Indexed: 11/28/2022]
Abstract
Due to growing antibiotic resistance, pneumonia caused by Pseudomonas aeruginosa is a major threat to human health and is driving the development of novel anti-infectious agents. Preventively or curatively administered pathogen-specific therapeutic antibodies (Abs) have several advantages, including a low level of toxicity and a unique pharmacological profile. At present, most Abs against respiratory infections are administered parenterally; this may not be optimal for therapeutics that have to reach the lungs to be effective. Although the airways constitute a logical delivery route for biologics designed to treat respiratory diseases, there are few scientific data on the advantages or disadvantages of this route in the context of pneumonia treatment. The objective of the present study was to evaluate the efficacy and fate of an anti-P. aeruginosa Ab targeting pcrV (mAb166) as a function of the administration route during pneumonia. The airway-administered mAb166 displayed a favorable pharmacokinetic profile during the acute phase of the infection, and was associated with greater protection (relative to other delivery routes) of infected animals. Airway administration was associated with lower levels of lung inflammation, greater bacterial clearance, and recruitment of neutrophils in the airways. In conclusion, the present study is the first to have compared the pharmacokinetics and efficacy of an anti-infectious Ab administered by different routes in an animal model of pneumonia. Our findings suggest that local delivery to the airways is associated with a more potent anti-bacterial response (relative to parenteral administration), and thus open up new perspectives for the prevention and treatment of pneumonia with Abs.
Collapse
Affiliation(s)
- Thomas Sécher
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | - Emilie Dalonneau
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | - Marion Ferreira
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France; CHRU de Tours, Département de Pneumologie et d'exploration respiratoire fonctionnelle, F-37032 Tours, France
| | - Christelle Parent
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | | | - Gilles Paintaud
- Université de Tours, GICC, PATCH Team, F-37032 Tours, France; CHRU de Tours, Laboratoire de Pharmacologie-Toxicologie, F-37032 Tours, France
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France
| | - Nathalie Heuzé-Vourc'h
- INSERM, Centre d'Etude des Pathologies Respiratoires, U1100, F-37032 Tours, France; Université de Tours, F-37032 Tours, France.
| |
Collapse
|
27
|
Gulin-Sarfraz T, Jonasson S, Wigenstam E, von Haartman E, Bucht A, Rosenholm JM. Feasibility Study of Mesoporous Silica Particles for Pulmonary Drug Delivery: Therapeutic Treatment with Dexamethasone in a Mouse Model of Airway Inflammation. Pharmaceutics 2019; 11:pharmaceutics11040149. [PMID: 30939753 PMCID: PMC6523761 DOI: 10.3390/pharmaceutics11040149] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/12/2019] [Accepted: 03/22/2019] [Indexed: 12/15/2022] Open
Abstract
Diseases in the respiratory tract rank among the leading causes of death in the world, and thus novel and optimized treatments are needed. The lungs offer a large surface for drug absorption, and the inhalation of aerosolized drugs are a well-established therapeutic modality for local treatment of lung conditions. Nanoparticle-based drug delivery platforms are gaining importance for use through the pulmonary route. By using porous carrier matrices, higher doses of especially poorly soluble drugs can be administered locally, reducing their side effects and improving their biodistribution. In this study, the feasibility of mesoporous silica particles (MSPs) as carriers for anti-inflammatory drugs in the treatment of airway inflammation was investigated. Two different sizes of particles on the micron and nanoscale (1 µm and 200 nm) were produced, and were loaded with dexamethasone (DEX) to a loading degree of 1:1 DEX:MSP. These particles were further surface-functionalized with a polyethylene glycol–polyethylene imine (PEG–PEI) copolymer for optimal aqueous dispersibility. The drug-loaded particles were administered as an aerosol, through inhalation to two different mice models of neutrophil-induced (by melphalan or lipopolysaccharide) airway inflammation. The mice received treatment with either DEX-loaded MSPs or, as controls, empty MSPs or DEX only; and were evaluated for treatment effects 24 h after exposure. The results show that the MEL-induced airway inflammation could be treated by the DEX-loaded MSPs to the same extent as free DEX. Interestingly, in the case of LPS-induced inflammation, even the empty MSPs significantly down-modulated the inflammatory response. This study highlights the potential of MSPs as drug carriers for the treatment of diseases in the airways.
Collapse
Affiliation(s)
- Tina Gulin-Sarfraz
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland.
- School of Pharmacy, University of Oslo, 0371 Oslo, Norway.
| | - Sofia Jonasson
- CBRN Defence and Security, Swedish Defence Research Agency, 90182 Umeå, Sweden.
| | - Elisabeth Wigenstam
- CBRN Defence and Security, Swedish Defence Research Agency, 90182 Umeå, Sweden.
| | - Eva von Haartman
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland.
| | - Anders Bucht
- CBRN Defence and Security, Swedish Defence Research Agency, 90182 Umeå, Sweden.
- Department of Public Health and Clinical Medicine, Unit of Respiratory Medicine, Umeå University, 90182 Umeå, Sweden.
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland.
| |
Collapse
|
28
|
Characterization of Host Responses during Pseudomonas aeruginosa Acute Infection in the Lungs and Blood and after Treatment with the Synthetic Immunomodulatory Peptide IDR-1002. Infect Immun 2018; 87:IAI.00661-18. [PMID: 30323028 PMCID: PMC6300642 DOI: 10.1128/iai.00661-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/02/2018] [Indexed: 12/26/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes nosocomial pneumonia and infects patients with cystic fibrosis. P. aeruginosa lung infections are difficult to treat due to bacterial resistance to antibiotics, and strains with multidrug resistance are becoming more prevalent. Pseudomonas aeruginosa is an opportunistic pathogen that causes nosocomial pneumonia and infects patients with cystic fibrosis. P. aeruginosa lung infections are difficult to treat due to bacterial resistance to antibiotics, and strains with multidrug resistance are becoming more prevalent. Here, we examined the use of a small host defense peptide, innate defense regulator 1002 (IDR-1002), in an acute P. aeruginosa lung infection in vivo. IDR-1002 significantly reduced the bacterial burden in bronchoalveolar lavage fluid (BALF), as well as MCP-1 in BALF and serum, KC in serum, and interleukin 6 (IL-6) in BALF. Transcriptome sequencing (RNA-Seq) was conducted on lungs and whole blood, and the effects of P. aeruginosa, IDR-1002, and the combination of P. aeruginosa and IDR-1002 were evaluated. Differential gene expression analysis showed that P. aeruginosa increased multiple inflammatory and innate immune pathways, as well as affected hemostasis, matrix metalloproteinases, collagen biosynthesis, and various metabolism pathways in the lungs and/or blood. Infected mice treated with IDR-1002 had significant changes in gene expression compared to untreated infected mice, with fewer differentially expressed genes associated with the inflammatory and innate immune responses to microbial infection, and treatment also affected morphogenesis, certain metabolic pathways, and lymphocyte activation. Overall, these results showed that IDR-1002 was effective in treating P. aeruginosa acute lung infections and associated inflammation.
Collapse
|
29
|
Nagre N, Cong X, Terrazas C, Pepper I, Schreiber JM, Fu H, Sill JM, Christman JW, Satoskar AR, Zhao X. Inhibition of Macrophage Complement Receptor CRIg by TRIM72 Polarizes Innate Immunity of the Lung. Am J Respir Cell Mol Biol 2018; 58:756-766. [PMID: 29268030 PMCID: PMC6002657 DOI: 10.1165/rcmb.2017-0236oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/22/2017] [Indexed: 12/13/2022] Open
Abstract
The complement system plays a critical role in immune responses against pathogens. However, its identity and regulation in the lung are not fully understood. This study aimed to explore the role of tripartite motif protein (TRIM) 72 in regulating complement receptor (CR) of the Ig superfamily (CRIg) in alveolar macrophage (AM) and innate immunity of the lung. Imaging, absorbance quantification, and flow cytometry were used to evaluate in vitro and in vivo AM phagocytosis with normal, or altered, TRIM72 expression. Pulldown, coimmunoprecipitation, and gradient binding assays were applied to examine TRIM72 and CRIg interaction. A pneumonia model was established by intratracheal injection of Pseudomonas aeruginosa. Mortality, lung bacterial burden, and cytokine levels in BAL fluid and lung tissues were examined. Our data show that TRIM72 inhibited CR-mediated phagocytosis, and release of TRIM72 inhibition led to increased AM phagocytosis. Biochemical assays identified CRIg as a binding partner of TRIM72, and TRIM72 inhibited formation of the CRIg-phagosome. Genetic ablation of TRIM72 led to improved pathogen clearance, reduced cytokine storm, and improved survival in murine models of severe pneumonia, specificity of which was confirmed by adoptive transfer of wild-type or TRIM72KO AMs to AM-depleted TRIM72KO mice. TRIM72 overexpression promoted bacteria-induced NF-κB activation in murine alveolar macrophage cells. Our data revealed a quiescent, noninflammatory bacterial clearance mechanism in the lung via AM CRIg, which is suppressed by TRIM72. In vivo data suggest that targeted suppression of TRIM72 in AM may be an effective measure to treat fatal pulmonary bacterial infections.
Collapse
Affiliation(s)
- Nagaraja Nagre
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Xiaofei Cong
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - César Terrazas
- Departments of Pathology and Microbiology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ian Pepper
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - John M. Schreiber
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Hongyun Fu
- Division of Community Health and Research, Pediatrics Department, Eastern Virginia Medical School, Norfolk, Virginia
| | - Joshua M. Sill
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, Virginia; and
| | - John W. Christman
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Abhay R. Satoskar
- Departments of Pathology and Microbiology, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Xiaoli Zhao
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| |
Collapse
|
30
|
Boucontet L, Passoni G, Thiry V, Maggi L, Herbomel P, Levraud JP, Colucci-Guyon E. A Model of Superinfection of Virus-Infected Zebrafish Larvae: Increased Susceptibility to Bacteria Associated With Neutrophil Death. Front Immunol 2018; 9:1084. [PMID: 29881380 PMCID: PMC5976802 DOI: 10.3389/fimmu.2018.01084] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 05/01/2018] [Indexed: 12/17/2022] Open
Abstract
Enhanced susceptibility to bacterial infection in the days following an acute virus infection such as flu is a major clinical problem. Mouse models have provided major advances in understanding viral-bacterial superinfections, yet interactions of the anti-viral and anti-bacterial responses remain elusive. Here, we have exploited the transparency of zebrafish to study how viral infections can pave the way for bacterial co-infections. We have set up a zebrafish model of sequential viral and bacterial infection, using sublethal doses of Sindbis virus and Shigella flexneri bacteria. This virus induces a strong type I interferons (IFN) response, while the bacterium induces a strong IL1β and TNFα-mediated inflammatory response. We found that virus-infected zebrafish larvae showed an increased susceptibility to bacterial infection. This resulted in the death with concomitant higher bacterial burden of the co-infected fish compared to the ones infected with bacteria only. By contrast, infecting with bacteria first and virus second did not lead to increased mortality or microbial burden. By high-resolution live imaging, we showed that neutrophil survival was impaired in Sindbis-then-Shigella co-infected fish. The two types of cytokine responses were strongly induced in co-infected fish. In addition to type I IFN, expression of the anti-inflammatory cytokine IL10 was induced by viral infection before bacterial superinfection. Collectively, these observations suggest the zebrafish larva as a useful animal model to address mechanisms underlying increased bacterial susceptibility upon viral infection.
Collapse
Affiliation(s)
- Laurent Boucontet
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Paris, France.,CNRS UMR 3738, Paris, France
| | - Gabriella Passoni
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Paris, France.,CNRS UMR 3738, Paris, France
| | - Valéry Thiry
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Paris, France.,CNRS UMR 3738, Paris, France
| | - Ludovico Maggi
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Paris, France.,CNRS UMR 3738, Paris, France
| | - Philippe Herbomel
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Paris, France.,CNRS UMR 3738, Paris, France
| | - Jean-Pierre Levraud
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Paris, France.,CNRS UMR 3738, Paris, France
| | - Emma Colucci-Guyon
- Institut Pasteur, Unité Macrophages et Développement de l'Immunité, Paris, France.,CNRS UMR 3738, Paris, France
| |
Collapse
|
31
|
Pan LL, Deng YY, Wang R, Wu C, Li J, Niu W, Yang Q, Bhatia M, Gudmundsson GH, Agerberth B, Diana J, Sun J. Lactose Induces Phenotypic and Functional Changes of Neutrophils and Macrophages to Alleviate Acute Pancreatitis in Mice. Front Immunol 2018; 9:751. [PMID: 29719535 PMCID: PMC5913286 DOI: 10.3389/fimmu.2018.00751] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 03/26/2018] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is one common clinical acute abdominal disease, for which specific pharmacological or nutritional therapies remain elusive. Lactose, a macronutrient and an inducer of host innate immune responses, possesses immune modulatory functions. The current study aimed to investigate potential modulatory effects of lactose and the interplay between the nutrient and pancreatic immunity during experimentally induced AP in mice. We found that either prophylactic or therapeutic treatment of lactose time-dependently reduced the severity of AP, as evidenced by reduced pancreatic edema, serum amylase levels, and pancreatic myeloperoxidase activities, as well as by histological examination of pancreatic damage. Overall, lactose promoted a regulatory cytokine milieu in the pancreas and reduced infiltration of inflammatory neutrophils and macrophages. On acinar cells, lactose was able to suppress caerulein-induced inflammatory signaling pathways and to suppress chemoattractant tumor necrosis factor (TNF)-α and monocyte chemotactic protein-1 production. Additionally, lactose acted on pancreas-infiltrated macrophages, increasing interleukin-10 and decreasing tumor necrosis factor alpha production. Notably, lactose treatment reversed AP-associated infiltration of activated neutrophils. Last, the effect of lactose on neutrophil infiltration was mimicked by a galectin-3 antagonist, suggesting a potential endogenous target of lactose. Together, the current study demonstrates an immune regulatory effect of lactose to alleviate AP and suggests its potential as a convenient, value-added therapeutic macronutrient to control AP, and lower the risk of its systemic complications.
Collapse
Affiliation(s)
- Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi, China
| | - Yuan-Yuan Deng
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ruxing Wang
- School of Medicine, Jiangnan University, Wuxi, China
- Wuxi People’s Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Chengfei Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wenying Niu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qin Yang
- School of Medicine, Jiangnan University, Wuxi, China
| | - Madhav Bhatia
- Inflammation Research Group, Department of Pathology, University of Otago, Christchurch, New Zealand
| | | | - Birgitta Agerberth
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Julien Diana
- Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 1151, Institute Necker-Enfants Malades (INEM), Centre National de la Recherche Scienctifique, Unité 8253, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- Nutrition and Immunology Laboratory, School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
32
|
Aoyagi T, Newstead MW, Zeng X, Nanjo Y, Peters-Golden M, Kaku M, Standiford TJ. Interleukin-36γ and IL-36 receptor signaling mediate impaired host immunity and lung injury in cytotoxic Pseudomonas aeruginosa pulmonary infection: Role of prostaglandin E2. PLoS Pathog 2017; 13:e1006737. [PMID: 29166668 PMCID: PMC5718565 DOI: 10.1371/journal.ppat.1006737] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Revised: 12/06/2017] [Accepted: 11/07/2017] [Indexed: 12/21/2022] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative pathogen that can lead to severe infection associated with lung injury and high mortality. The interleukin (IL)-36 cytokines (IL-36α, IL-36β and IL-36γ) are newly described IL-1 like family cytokines that promote inflammatory response via binding to the IL-36 receptor (IL-36R). Here we investigated the functional role of IL-36 cytokines in the modulating of innate immune response against P. aeruginosa pulmonary infection. The intratracheal administration of flagellated cytotoxic P. aeruginosa (ATCC 19660) upregulated IL-36α and IL-36γ, but not IL-36β, in the lungs. IL-36α and IL-36γ were expressed in pulmonary macrophages (PMs) and alveolar epithelial cells in response to P. aeruginosa in vitro. Mortality after bacterial challenge in IL-36 receptor deficient (IL-36R-/-) mice and IL-36γ deficient (IL-36γ-/-) mice, but not IL-36α deficient mice, was significantly lower than that of wild type mice. Decreased mortality in IL-36R-/- mice and IL-36γ-/- mice was associated with reduction in bacterial burden in the alveolar space, bacterial dissemination, production of inflammatory cytokines and lung injury, without changes in lung leukocyte influx. Interestingly, IL-36γ enhanced the production of prostaglandin E2 (PGE2) during P. aeruginosa infection in vivo and in vitro. Treatment of PMs with recombinant IL-36γ resulted in impaired bacterial killing via PGE2 and its receptor; EP2. P. aeruginosa infected EP2 deficient mice or WT mice treated with a COX-2-specific inhibitor showed decreased bacterial burden and dissemination, but no change in lung injury. Finally, we observed an increase in IL-36γ, but not IL-36α, in the airspace and plasma of patients with P. aeruginosa-induced acute respiratory distress syndrome. Thus, IL-36γ and its receptor signal not only impaired bacterial clearance in a possible PGE2 dependent fashion but also mediated lung injury during P. aeruginosa infection.
Collapse
Affiliation(s)
- Tetsuji Aoyagi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Infection Control and Laboratory Diagnostics, Internal Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- * E-mail:
| | - Michael W. Newstead
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Xianying Zeng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Yuta Nanjo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Microbiology and Infectious Diseases, Toho University School of Medicine, Tokyo, Japan
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Mitsuo Kaku
- Department of Infection Control and Laboratory Diagnostics, Internal Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Theodore J. Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
33
|
He Y, Wu C, Li J, Li H, Sun Z, Zhang H, de Vos P, Pan LL, Sun J. Inulin-Type Fructans Modulates Pancreatic-Gut Innate Immune Responses and Gut Barrier Integrity during Experimental Acute Pancreatitis in a Chain Length-Dependent Manner. Front Immunol 2017; 8:1209. [PMID: 29018453 PMCID: PMC5622924 DOI: 10.3389/fimmu.2017.01209] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/12/2017] [Indexed: 12/11/2022] Open
Abstract
Acute pancreatitis (AP) is a common abdominal inflammatory disorder and one of the leading causes of hospital admission for gastrointestinal disorders. No specific pharmacological or nutritional therapy is available but highly needed. Inulin-type fructans (ITFs) are capable of modifying gut immune and barrier homeostasis in a chemistry-dependent manner and hence potentially applicable for managing AP, but their efficacy in AP has not been demonstrated yet. The current study aimed to examine and compare modulatory effects of ITFs with different degrees of fermentability on pancreatic-gut immunity and barrier function during experimentally induced AP in mice. BALB/c mice were fed short (I)- or long (IV)-chain ITFs supplemented diets for up to 3 days before AP induction by caerulein. Attenuating effects on AP development were stronger with ITF IV than with ITF I. We found that long-chain ITF IV attenuated the severity of AP, as evidenced by reduced serum amylase levels, lipase levels, pancreatic myeloperoxidase activity, pancreatic edema, and histological examination demonstrating reduced pancreatic damage. Short-chain ITF I demonstrated only partial protective effects. Both ITF IV and ITF I modulated AP-associated systemic cytokine levels. ITF IV but not ITF I restored AP-associated intestinal barrier dysfunction by upregulating colonic tight junction modulatory proteins, antimicrobial peptides, and improved general colonic histology. Additionally, differential modulatory effects of ITF IV and ITF I were observed on pancreatic and gut immunity: ITF IV supplementation prevented innate immune cell infiltration in the pancreas and colon and tissue cytokine production. Similar effects were only observed in the gut with ITF I and not in the pancreas. Lastly, ITF IV but not ITF I downregulated AP-triggered upregulation of IL-1 receptor-associated kinase 4 (IRAK-4) and phosphor-c-Jun N-terminal kinase (p-JNK), and a net decrease of phosphor-nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 (p-NF-κB p65) nuclear translocation and activation in the pancreas. Our findings demonstrate a clear chain length-dependent effect of inulin on AP. The attenuating effects are caused by modulating effects of long-chain inulin on the pancreatic-gut immunity via the pancreatic IRAK-4/p-JNK/p-NF-κBp65 signaling pathway and on prevention of disruption of the gut barrier.
Collapse
Affiliation(s)
- Yue He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Chengfei Wu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hongli Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Zhenghua Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Paul de Vos
- Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi, China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
34
|
Wilson ZS, Ahn LB, Serratelli WS, Belley MD, Lomas-Neira J, Sen M, Lefort CT. Activated β 2 Integrins Restrict Neutrophil Recruitment during Murine Acute Pseudomonal Pneumonia. Am J Respir Cell Mol Biol 2017; 56:620-627. [PMID: 28157452 DOI: 10.1165/rcmb.2016-0215oc] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Rapid neutrophil recruitment is critical for the efficient clearance of bacterial pathogens from the lungs. Although β2 integrins and their activation are required for neutrophil recruitment from postcapillary venules of the systemic circulation into inflamed tissues, the involvement of integrins in neutrophil recruitment in response to respiratory infection varies among bacterial pathogens. For stimuli eliciting β2 integrin-dependent neutrophil influx, including Pseudomonas aeruginosa, it remains unclear whether the activation of β2 integrins is an essential step in this process. In the current study, we analyze neutrophil trafficking within the lungs of mice infected with Pseudomonas aeruginosa and evaluate the role of β2 integrin activation through genetic deletion of talin-1 or Kindlin-3 or by pharmacological inhibition of high-affinity β2 integrins using a small molecule allosteric antagonist. We observe that attenuation of high-affinity β2 integrins leads to an enhancement of neutrophil emigration into lung interstitium and airspaces. Neutrophil effector functions, including the production of reactive oxygen species and the phagocytosis of bacteria, are only partially dependent on high-affinity β2 integrins. These results reveal a mechanism by which activated β2 integrins limit neutrophil entry into the lung tissue and airspaces during acute pseudomonal pneumonia and suggest potential strategies for modulating neutrophil-mediated host defense.
Collapse
Affiliation(s)
- Zachary S Wilson
- 1 Division of Surgical Research, Department of Surgery, and.,2 Graduate Program in Pathobiology and
| | - Lawrence B Ahn
- 1 Division of Surgical Research, Department of Surgery, and
| | | | - Matthew D Belley
- 4 Department of Radiation Oncology, Rhode Island Hospital, Providence, Rhode Island.,3 Warren Alpert Medical School, Brown University, Providence, Rhode Island.,5 Department of Physics, University of Rhode Island, Kingston, Rhode Island; and
| | | | - Mehmet Sen
- 6 Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Craig T Lefort
- 1 Division of Surgical Research, Department of Surgery, and.,3 Warren Alpert Medical School, Brown University, Providence, Rhode Island
| |
Collapse
|
35
|
Sun L, Pham TT, Cornell TT, McDonough KL, McHugh WM, Blatt NB, Dahmer MK, Shanley TP. Myeloid-Specific Gene Deletion of Protein Phosphatase 2A Magnifies MyD88- and TRIF-Dependent Inflammation following Endotoxin Challenge. THE JOURNAL OF IMMUNOLOGY 2016; 198:404-416. [PMID: 27872207 DOI: 10.4049/jimmunol.1600221] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 10/21/2016] [Indexed: 12/23/2022]
Abstract
Protein phosphatase 2A (PP2A) is a member of the intracellular serine/threonine phosphatases. Innate immune cell activation triggered by pathogen-associated molecular patterns is mediated by various protein kinases, and PP2A plays a counter-regulatory role by deactivating these kinases. In this study, we generated a conditional knockout of the α isoform of the catalytic subunit of PP2A (PP2ACα). After crossing with myeloid-specific cre-expressing mice, effective gene knockout was achieved in various myeloid cells. The myeloid-specific knockout mice (lyM-PP2Afl/fl) showed higher mortality in response to endotoxin challenge and bacterial infection. Upon LPS challenge, serum levels of TNF-α, KC, IL-6, and IL-10 were significantly increased in lyM-PP2Afl/fl mice, and increased phosphorylation was observed in MAPK pathways (p38, ERK, JNK) and the NF-κB pathway (IKKα/β, NF-κB p65) in bone marrow-derived macrophages (BMDMs) from knockout mice. Heightened NF-κB activation was not associated with degradation of IκBα; instead, enhanced phosphorylation of the NF-κB p65 subunit and p38 phosphorylation-mediated TNF-α mRNA stabilization appear to contribute to the increased TNF-α expression. In addition, increased IL-10 expression appears to be due to PP2ACα-knockout-induced IKKα/β hyperactivation. Microarray experiments indicated that the Toll/IL-1R domain-containing adaptor inducing IFN-β/ TNFR-associated factor 3 pathway was highly upregulated in LPS-treated PP2ACα-knockout BMDMs, and knockout BMDMs had elevated IFN-α/β production compared with control BMDMs. Serum IFN-β levels from PP2ACα-knockout mice treated with LPS were also greater than those in controls. Thus, we demonstrate that PP2A plays an important role in regulating inflammation and survival in the setting of septic insult by targeting MyD88- and Toll/IL-1R domain-containing adaptor inducing IFN-β-dependent pathways.
Collapse
Affiliation(s)
- Lei Sun
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109;
| | - Tiffany T Pham
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Timothy T Cornell
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Kelli L McDonough
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Walker M McHugh
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Neal B Blatt
- Division of Pediatric Nephrology, Department of Pediatrics and Communicable Diseases, C.S. Mott Children's Hospital, University of Michigan Medical School, Ann Arbor, MI 48109; and
| | - Mary K Dahmer
- Division of Critical Care Medicine, Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Thomas P Shanley
- Department of Pediatrics, Lurie Children's Hospital of Chicago, Feinberg School of Medicine, Northwestern University, Evanston, IL 60611
| |
Collapse
|
36
|
Kamei A, Gao G, Neale G, Loh LN, Vogel P, Thomas PG, Tuomanen EI, Murray PJ. Exogenous remodeling of lung resident macrophages protects against infectious consequences of bone marrow-suppressive chemotherapy. Proc Natl Acad Sci U S A 2016; 113:E6153-E6161. [PMID: 27671632 PMCID: PMC5068317 DOI: 10.1073/pnas.1607787113] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Infection is the single greatest threat to survival during cancer chemotherapy because of depletion of bone marrow-derived immune cells. Phagocytes, especially neutrophils, are key effectors in immunity to extracellular pathogens, which has limited the development of new approaches to protect patients with cancer and chemotherapy-induced neutropenia. Using a model of vaccine-induced protection against lethal Pseudomonas aeruginosa pneumonia in the setting of chemotherapy-induced neutropenia, we found a population of resident lung macrophages in the immunized lung that mediated protection in the absence of neutrophils, bone marrow-derived monocytes, or antibodies. These vaccine-induced macrophages (ViMs) expanded after immunization, locally proliferated, and were closely related to alveolar macrophages (AMs) by surface phenotype and gene expression profiles. By contrast to AMs, numbers of ViMs were stable through chemotherapy, showed enhanced phagocytic activity, and prolonged survival of neutropenic mice from lethal P. aeruginosa pneumonia upon intratracheal adoptive transfer. Thus, induction of ViMs by tissue macrophage remodeling may become a framework for new strategies to activate immune-mediated reserves against infection in immunocompromised hosts.
Collapse
Affiliation(s)
- Akinobu Kamei
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105; Department of Pediatrics, Keio University School of Medicine, Tokyo 160-8582, Japan;
| | - Geli Gao
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Geoffrey Neale
- Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Lip Nam Loh
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Peter Vogel
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Elaine I Tuomanen
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Peter J Murray
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN 38105; Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| |
Collapse
|
37
|
Peñaloza HF, Schultz BM, Nieto PA, Salazar GA, Suazo I, Gonzalez PA, Riedel CA, Alvarez-Lobos MM, Kalergis AM, Bueno SM. Opposing roles of IL-10 in acute bacterial infection. Cytokine Growth Factor Rev 2016; 32:17-30. [PMID: 27522641 DOI: 10.1016/j.cytogfr.2016.07.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 07/14/2016] [Indexed: 12/16/2022]
Abstract
Interleukin-10 (IL-10) is recognized as an anti-inflammatory cytokine that downmodulates inflammatory immune responses at multiple levels. In innate cells, production of this cytokine is usually triggered after pathogen recognition receptor (PRR) engagement by pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patters (DAMPs), as well as by other soluble factors. Importantly, IL-10 is frequently secreted during acute bacterial infections and has been described to play a key role in infection resolution, although its effects can significantly vary depending on the infecting bacterium. While the production of IL-10 might favor host survival in some cases, it may also result harmful for the host in other circumstances, as it can prevent appropriate bacterial clearance. In this review we discuss the role of IL-10 in bacterial clearance and propose that this cytokine is required to recover from infection caused by extracellular or highly pro-inflammatory bacteria. Altogether, we propose that IL-10 drives excessive suppression of the immune response upon infection with intracellular bacteria or in non-inflammatory bacterial infections, which ultimately favors bacterial persistence and dissemination within the host. Thus, the nature of the bacterium causing infection is an important factor that needs to be taken into account when considering new immunotherapies that consist on the modulation of inflammation, such as IL-10. Indeed, induction of this cytokine may significantly improve the host's immune response to certain bacteria when antibiotics are not completely effective.
Collapse
Affiliation(s)
- Hernán F Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Barbara M Schultz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Pamela A Nieto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Geraldyne A Salazar
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Isidora Suazo
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Pablo A Gonzalez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello, Chile
| | - Manuel M Alvarez-Lobos
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile; INSERM U1064, Nantes, France
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Pontificia Universidad Católica de Chile, Chile; INSERM U1064, Nantes, France.
| |
Collapse
|
38
|
Immunogenicity and protective efficacy of Pseudomonas aeruginosa type a and b flagellin vaccines in a burned mouse model. Mol Immunol 2016; 74:71-81. [DOI: 10.1016/j.molimm.2016.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 04/07/2016] [Accepted: 04/14/2016] [Indexed: 12/31/2022]
|
39
|
Peñaloza HF, Nieto PA, Muñoz-Durango N, Salazar-Echegarai FJ, Torres J, Parga MJ, Alvarez-Lobos M, Riedel CA, Kalergis AM, Bueno SM. Interleukin-10 plays a key role in the modulation of neutrophils recruitment and lung inflammation during infection by Streptococcus pneumoniae. Immunology 2015; 146:100-12. [PMID: 26032199 DOI: 10.1111/imm.12486] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 05/03/2015] [Accepted: 05/27/2015] [Indexed: 12/29/2022] Open
Abstract
Streptococcus pneumoniae is a major aetiological agent of pneumonia worldwide, as well as otitis media, sinusitis, meningitis and sepsis. Recent reports have suggested that inflammation of lungs due to S. pneumoniae infection promotes bacterial dissemination and severe disease. However, the contribution of anti-inflammatory molecules to the pathogenesis of S. pneumoniae remains unknown. To elucidate whether the production of the anti-inflammatory cytokine interleukin-10 (IL-10) is beneficial or detrimental for the host during pneumococcal pneumonia, we performed S. pneumoniae infections in mice lacking IL-10 (IL-10(-/-) mice). The IL-10(-/-) mice showed increased mortality, higher expression of pro-inflammatory cytokines, and an exacerbated recruitment of neutrophils into the lungs after S. pneumoniae infection. However, IL-10(-/-) mice showed significantly lower bacterial loads in lungs, spleen, brain and blood, when compared with mice that produced this cytokine. Our results support the notion that production of IL-10 during S. pneumoniae infection modulates the expression of pro-inflammatory cytokines and the infiltration of neutrophils into the lungs. This feature of IL-10 is important to avoid excessive inflammation of tissues and to improve host survival, even though bacterial dissemination is less efficient in the absence of this cytokine.
Collapse
Affiliation(s)
- Hernán F Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pamela A Nieto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natalia Muñoz-Durango
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco J Salazar-Echegarai
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Javiera Torres
- Departamento de Anatomía Patológica, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María J Parga
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel Alvarez-Lobos
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas y Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Inmunología Clínica y Reumatología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,INSERM U1064, Nantes, France
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,INSERM U1064, Nantes, France
| |
Collapse
|
40
|
Taddonio MA, Dolgachev V, Bosmann M, Ward PA, Su G, Wang SC, Hemmila MR. Influence of lipopolysaccharide-binding protein on pulmonary inflammation in gram-negative pneumonia. Shock 2015; 43:612-9. [PMID: 25643011 PMCID: PMC4433570 DOI: 10.1097/shk.0000000000000349] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lipopolysaccharide-binding protein (LBP) is upregulated as part of the acute-phase response. Lipopolysaccharide-binding protein has a known multifunctional role in potentiating the recognition, clearance, and killing of gram-negative bacteria. In a Klebsiella pneumonia model, we previously demonstrated that LBP gene-deficient mice (LBP-/-) mice experience increased mortality when compared with wild-type (Wt) mice (98% vs. 59%). We hypothesize that LBP is essential to bacterial clearance from the lung, and its absence leads to alteration of the pulmonary inflammatory response to pneumonia. Twelve- to 16-week-old female C57Bl/6 Wt mice and age-matched LBP-/- mice were administered 1 × 10(3) colony-forming units of Klebsiella pneumoniae by intratracheal injection. Animals were euthanized at 6, 12, 24, or 36 h after inoculation. Lung tissue and bronchoalveolar lavage samples were obtained. Lung homogenate samples were assayed to determine quantitative bacterial load per whole lung, proinflammatory cytokine concentrations, myeloperoxidase activity, and assessment of pulmonary leukocyte populations. In vitro production of inflammatory mediators were also assayed after LPS stimulation of peritoneal macrophages isolated from Wt, Toll-like receptor 4 (TLR4)-deficient, and LBP-/- mice. The LBP-/- mice demonstrated significantly elevated levels of bacteria in the lung at 24 and 36 h when compared with Wt controls. The average lung levels of proinflammatory cytokines interleukin-1β (IL-1β), IL-6, keratinocyte-derived chemokine, and macrophage-inflammatory protein-2 were greater in the LBP mice and remained elevated longer when compared with those in the Wt mice. Myeloperoxidase activity, an indicator of neutrophil content, was significantly increased at time 36 h in the LBP mice. After in vitro stimulation of peritoneal macrophages with LPS, production of IL-1β, IL-6, IL-10, keratinocyte-derived chemokine, and macrophage-inflammatory protein-1α were suppressed in LBP and TLR4-deficient mice compared with that in Wt. Absence of a functional LBP-/- gene results in diminished clearance of gram-negative bacteria from the pulmonary system. Failure to recognize and clear gram-negative bacteria via the LBP/TLR4 axis results in an initial delayed inflammatory response. This delay in LBP-/- mice is followed by excessive amplification and prolonged elevation of proinflammatory mediators and neutrophil sequestration within the lungs.
Collapse
Affiliation(s)
- Michael A. Taddonio
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Vladislav Dolgachev
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Markus Bosmann
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Peter A. Ward
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Grace Su
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Stewart C. Wang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Mark R. Hemmila
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
41
|
Huang R, Zhong T, Wu H. Quercetin protects against lipopolysaccharide-induced acute lung injury in rats through suppression of inflammation and oxidative stress. Arch Med Sci 2015; 11:427-32. [PMID: 25995762 PMCID: PMC4424260 DOI: 10.5114/aoms.2015.50975] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 03/22/2013] [Accepted: 05/20/2013] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Acute lung injury (ALI) is an acute inflammatory disease characterized by excess production of inflammatory factors in lung tissue. Quercetin, a herbal flavonoid, exhibits anti-inflammatory and anti-oxidative properties. This study was performed to assess the effects of quercetin on lipopolysaccharide (LPS)-induced ALI. MATERIAL AND METHODS Sprague-Dawley rats were randomly divided into 3 groups: the control group (saline alone), the LPS group challenged with LPS (Escherichia coli 026:B6; 100 µg/kg), and the quercetin group pretreated with quercetin (50 mg/kg, by gavage) 1 h before LPS challenge. Bronchoalveolar lavage fluid (BALF) samples and lung tissues were collected 6 h after LPS administration. Histopathological and biochemical parameters were measured. RESULTS The LPS treatment led to increased alveolar wall thickening and cellular infiltration in the lung, which was markedly prevented by quercetin pretreatment. Moreover, quercetin significantly (p < 0.05) attenuated the increase in the BALF protein level and neutrophil count and lung wet/dry weight ratio and myeloperoxidase activity in LPS-challenged rats. The LPS exposure evoked a 4- to 5-fold rise in BALF levels of tumor necrosis factor-α and interleukin-6, which was significantly (p < 0.05) counteracted by quercetin pretreatment. Additionally, quercetin significantly (p < 0.05) suppressed the malondialdehyde level and increased the activities of superoxide dismutase, catalase, and glutathione peroxidase in the lung of LPS-treated rats. CONCLUSIONS Quercetin pretreatment effectively ameliorates LPS-induced ALI, largely through suppression of inflammation and oxidative stress, and may thus have therapeutic potential in the prevention of this disease.
Collapse
Affiliation(s)
- Risheng Huang
- Department of Thoracic Surgery, Wenzhou Central Hospital, Wenzhou, China
| | - Tian Zhong
- Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, China
| | - Hao Wu
- Department of Clinical Laboratory, Wenzhou Central Hospital, Wenzhou, China
| |
Collapse
|
42
|
Maile R, Jones S, Pan Y, Zhou H, Jaspers I, Peden DB, Cairns BA, Noah TL. Association between early airway damage-associated molecular patterns and subsequent bacterial infection in patients with inhalational and burn injury. Am J Physiol Lung Cell Mol Physiol 2015; 308:L855-60. [PMID: 25770180 DOI: 10.1152/ajplung.00321.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/10/2015] [Indexed: 01/24/2023] Open
Abstract
Bacterial infection is a major cause of morbidity affecting outcome following burn and inhalation injury. While experimental burn and inhalation injury animal models have suggested that mediators of cell damage and inflammation increase the risk of infection, few studies have been done on humans. This is a prospective, observational study of patients admitted to the North Carolina Jaycee Burn Center at the University of North Carolina who were intubated and on mechanical ventilation for treatment of burn and inhalational injury. Subjects were enrolled over a 2-yr period and followed till discharge or death. Serial bronchial washings from clinically indicated bronchoscopies were collected and analyzed for markers of tissue injury and inflammation. These include damage-associated molecular patterns (DAMPs) such as hyaluronic acid (HA), double-stranded DNA (dsDNA), heat-shock protein 70 (HSP-70), and high-mobility group protein B-1 (HMGB-1). The study population was comprised of 72 patients who had bacterial cultures obtained for clinical indications. Elevated HA, dsDNA, and IL-10 levels in bronchial washings obtained early (the first 72 h after injury) were significantly associated with positive bacterial respiratory cultures obtained during the first 14 days postinjury. Independent of initial inhalation injury severity and extent of surface burn, elevated levels of HA dsDNA and IL-10 in the central airways obtained early after injury are associated with subsequent positive bacterial respiratory cultures in patients intubated after acute burn/inhalation injury.
Collapse
Affiliation(s)
- Robert Maile
- Department of Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; North Carolina Jaycee Burn Center, Chapel Hill, North Carolina;
| | - Samuel Jones
- Department of Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; North Carolina Jaycee Burn Center, Chapel Hill, North Carolina
| | - Yinghao Pan
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Haibo Zhou
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ilona Jaspers
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - David B Peden
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Bruce A Cairns
- Department of Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; North Carolina Jaycee Burn Center, Chapel Hill, North Carolina
| | - Terry L Noah
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| |
Collapse
|
43
|
Baral P, Batra S, Zemans RL, Downey GP, Jeyaseelan S. Divergent functions of Toll-like receptors during bacterial lung infections. Am J Respir Crit Care Med 2015; 190:722-32. [PMID: 25033332 DOI: 10.1164/rccm.201406-1101pp] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Lower respiratory tract infections caused by bacteria are a major cause of death in humans irrespective of sex, race, or geography. Indeed, accumulated data indicate greater mortality and morbidity due to these infections than cancer, malaria, or HIV infection. Successful recognition of, followed by an appropriate response to, bacterial pathogens in the lungs is crucial for effective pulmonary host defense. Although the early recruitment and activation of neutrophils in the lungs is key in the response against invading microbial pathogens, other sentinels, such as alveolar macrophages, epithelial cells, dendritic cells, and CD4(+) T cells, also contribute to the elimination of the bacterial burden. Pattern recognition receptors, such as Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain-like receptors, are important for recognizing and responding to microbes during pulmonary infections. However, bacterial pathogens have acquired crafty evasive strategies to circumvent the pattern recognition receptor response and thus establish infection. Increased understanding of the function of TLRs and evasive mechanisms used by pathogens during pulmonary infection will deepen our knowledge of immunopathogenesis and is crucial for developing effective therapeutic and/or prophylactic measures. This review summarizes current knowledge of the multiple roles of TLRs in bacterial lung infections and highlights the mechanisms used by pathogens to modulate or interfere with TLR signaling in the lungs.
Collapse
Affiliation(s)
- Pankaj Baral
- 1 Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | | | | | | | | |
Collapse
|
44
|
Interleukin 10 overexpression alters survival in the setting of gram-negative pneumonia following lung contusion. Shock 2014; 41:301-10. [PMID: 24430542 DOI: 10.1097/shk.0000000000000123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Lung contusion injury produces a vulnerable window within the inflammatory defenses of the lung that predisposes the patient to pneumonia. Interleukin 10 (IL-10) is a known anti-inflammatory mediator produced by macrophages and capable of downregulating acute lung inflammation. We investigated the impact of increased levels of IL-10 within the lung on survival and the host response to trauma in the setting of lung contusion (LC) and gram-negative pneumonia. DESIGN A bitransgenic, tetracycline-inducible, lung-specific human IL-10 overexpression (IL-10 OE) mouse model and single transgenic (TG-) control mice were used. Mice underwent LC injury or sham injury (sham) at time -6 h. At time 0, animals were inoculated intratracheally with 500 colony-forming units of Klebsiella pneumoniae (pneu). Bronchoalveolar lavage fluid, lung tissue specimens, or purified macrophages were collected. Lung tissue and blood bacteria levels were quantified. Cytokine levels were assayed by enzyme-linked immunosorbent assay, and gene expression levels were evaluated by real-time polymerase chain reaction. Cell-type identification and quantification were done using real-time polymerase chain reaction and flow cytometry. MAIN RESULTS Interleukin 10 OE mice demonstrated decreased 5-day survival compared with TG- mice following LC + pneu (0 vs. 30%, P < 0.0001). Interleukin 10 OE mice had significantly higher lung bacteria counts (P = 0.02) and levels of bacteremia (P = 0.001) at 24 h. The IL-10 OE mice recruited more neutrophils into the alveoli as measured in bronchoalveolar lavage fluid compared with TG- mice. Alveolar macrophages from IL-10 OE mice displayed increased alternative activation (M2 macrophages, P = 0.046), whereas macrophages from TG- mice exhibited classic activation (M1 macrophages) and much higher intracellular bacterial killing potential (P = 0.03). Interleukin 6, keratinocyte-derived chemokine, and macrophage inflammatory protein 2 levels were significantly elevated in IL-10 OE LC + pneu animals (P < 0.05). CONCLUSIONS Lung-specific IL-10 overexpression induces alternative activation of alveolar macrophages. This shift in macrophage phenotype decreases intracellular bacterial killing, resulting in a more pronounced bacteremia and accelerated mortality in a model of LC and pneumonia.
Collapse
|
45
|
Heat-shock response increases lung injury caused by Pseudomonas aeruginosa via an interleukin-10-dependent mechanism in mice. Anesthesiology 2014; 120:1450-62. [PMID: 24667831 DOI: 10.1097/aln.0000000000000235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND The heat-shock response (HSR) protects from insults, such as ischemia-reperfusion injury, by inhibiting signaling pathways activated by sterile inflammation. However, the mechanisms by which the HSR activation would modulate lung damage and host response to a bacterial lung infection remain unknown. METHODS HSR was activated with whole-body hyperthermia or by intraperitoneal geldanamycin in mice that had their lungs instilled with Pseudomonas aeruginosa 24 h later (at least six mice per experimental group). Four hours after instillation, lung endothelial and epithelial permeability, bacterial counts, protein levels in bronchoalveolar lavage fluid, and lung myeloperoxidase activity were measured. Mortality rate 24 h after P. aeruginosa instillation was recorded. The HSR effect on the release of interleukin-10 and killing of P. aeruginosa bacteria by a mouse alveolar macrophage cell line and on neutrophil phagocytosis was also examined. RESULTS HSR activation worsened lung endothelial (42%) and epithelial permeability (50%) to protein, decreased lung bacterial clearance (71%), and increased mortality (50%) associated with P. aeruginosa pneumonia, an effect that was not observed in heat-shock protein-72-null mice. HSR-mediated decrease in neutrophil phagocytosis (69%) and bacterial killing (38%) by macrophages was interleukin-10 dependent, a mechanism confirmed by increased lung bacterial clearance and decreased mortality (70%) caused by P. aeruginosa pneumonia in heat-shocked interleukin-10-null mice. CONCLUSIONS Prior HSR activation worsens lung injury associated with P. aeruginosa pneumonia in mice via heat-shock protein-72- and interleukin-10-dependent mechanisms. These results provide a novel mechanism for the immunosuppression observed after severe trauma that is known to activate HSR in humans.
Collapse
|
46
|
Coinfection with an intestinal helminth impairs host innate immunity against Salmonella enterica serovar Typhimurium and exacerbates intestinal inflammation in mice. Infect Immun 2014; 82:3855-66. [PMID: 24980971 DOI: 10.1128/iai.02023-14] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Salmonella enterica serovar Typhimurium is a Gram-negative food-borne pathogen that is a major cause of acute gastroenteritis in humans. The ability of the host to control such bacterial pathogens may be influenced by host immune status and by concurrent infections. Helminth parasites are of particular interest in this context because of their ability to modulate host immune responses and because their geographic distribution coincides with those parts of the world where infectious gastroenteritis is most problematic. To test the hypothesis that helminth infection may negatively regulate host mucosal innate immunity against bacterial enteropathogens, a murine coinfection model was established by using the intestinal nematode Heligmosomoides polygyrus and S. Typhimurium. We found that mice coinfected with S. Typhimurium and H. polygyrus developed more severe intestinal inflammation than animals infected with S. Typhimurium alone. The enhanced susceptibility to Salmonella-induced intestinal injury in coinfected mice was found to be associated with diminished neutrophil recruitment to the site of bacterial infection that correlated with decreased expression of the chemoattractants CXCL2/macrophage inflammatory protein 2 (MIP-2) and CXCL1/keratinocyte-derived chemokine (KC), poor control of bacterial replication, and exacerbated intestinal inflammation. The mechanism of helminth-induced inhibition of MIP-2 and KC expression involved interleukin-10 (IL-10) and, to a lesser extent, IL-4 and IL-13. Ly6G antibody-mediated depletion of neutrophils reproduced the adverse effects of H. polygyrus on Salmonella infection. Our results suggest that impaired neutrophil recruitment is an important contributor to the enhanced severity of Salmonella enterocolitis associated with helminth coinfection.
Collapse
|
47
|
Aggarwal NR, Tsushima K, Eto Y, Tripathi A, Mandke P, Mock JR, Garibaldi BT, Singer BD, Sidhaye VK, Horton MR, King LS, D'Alessio FR. Immunological priming requires regulatory T cells and IL-10-producing macrophages to accelerate resolution from severe lung inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2014; 192:4453-4464. [PMID: 24688024 PMCID: PMC4001810 DOI: 10.4049/jimmunol.1400146] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Overwhelming lung inflammation frequently occurs following exposure to both direct infectious and noninfectious agents and is a leading cause of mortality worldwide. In that context, immunomodulatory strategies may be used to limit severity of impending organ damage. We sought to determine whether priming the lung by activating the immune system, or immunological priming, could accelerate resolution of severe lung inflammation. We assessed the importance of alveolar macrophages, regulatory T cells, and their potential interaction during immunological priming. We demonstrate that oropharyngeal delivery of low-dose LPS can immunologically prime the lung to augment alveolar macrophage production of IL-10 and enhance resolution of lung inflammation induced by a lethal dose of LPS or by Pseudomonas bacterial pneumonia. IL-10-deficient mice did not achieve priming and were unable to accelerate lung injury resolution. Depletion of lung macrophages or regulatory T cells during the priming response completely abrogated the positive effect of immunological priming on resolution of lung inflammation and significantly reduced alveolar macrophage IL-10 production. Finally, we demonstrated that oropharyngeal delivery of synthetic CpG-oligonucleotides elicited minimal lung inflammation compared with low-dose LPS but nonetheless primed the lung to accelerate resolution of lung injury following subsequent lethal LPS exposure. Immunological priming is a viable immunomodulatory strategy used to enhance resolution in an experimental acute lung injury model with the potential for therapeutic benefit against a wide array of injurious exposures.
Collapse
Affiliation(s)
- Neil R Aggarwal
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Kenji Tsushima
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Yoshiki Eto
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Ashutosh Tripathi
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Pooja Mandke
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Jason R Mock
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Brian T Garibaldi
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Venkataramana K Sidhaye
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Maureen R Horton
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Landon S King
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Franco R D'Alessio
- Division of Pulmonary and Critical Care Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21224
| |
Collapse
|
48
|
Yan X, Hegab AE, Endo J, Anzai A, Matsuhashi T, Katsumata Y, Ito K, Yamamoto T, Betsuyaku T, Shinmura K, Shen W, Vivier E, Fukuda K, Sano M. Lung Natural Killer Cells Play a Major Counter-Regulatory Role in Pulmonary Vascular Hyperpermeability After Myocardial Infarction. Circ Res 2014; 114:637-49. [DOI: 10.1161/circresaha.114.302625] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale
:
Natural killer (NK) cells are lymphocytes of the innate immune system that play specialized and niche-specific roles in distinct organs.
Objective
:
We investigated the possible function of NK cells in the pathogenesis of congestive heart failure after myocardial infarction.
Methods and Results
:
Depletion of NK cells from mice had little effect on cytokine expression (tumor necrosis factor-α, interleukin [IL]-6, and IL-1β), neutrophil and macrophage infiltration into infarcted myocardium, or left ventricular remodeling after myocardial infarction. However, these mice exhibited severe respiratory distress associated with protein-rich, high-permeability alveolar edema accompanied by neutrophil infiltration. In addition, there were 20-fold more NK cells in the mouse lungs than in heart, and these cells were accumulated around the vasculature. CD107a-positive and interferon-γ–positive cell populations were unchanged, whereas IL-10–positive populations increased. Adoptive transfer of NK cells from wild-type mice, but not from IL-10 knockout mice, into the NK cell–depleted mice rescued the respiratory phenotype. IL-1β–mediated dextran leakage from a lung endothelial cell monolayer was also blocked by coculture with NK cells from wild-type mice but not from IL-10 knockout mice.
Conclusions
:
This study is the first to identify a critical role for lung NK cells in protecting lung from the development of cardiogenic pulmonary edema after myocardial infarction.
Collapse
Affiliation(s)
- Xiaoxiang Yan
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Ahmed E. Hegab
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Jin Endo
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Atsushi Anzai
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Tomohiro Matsuhashi
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Yoshinori Katsumata
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Kentaro Ito
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Tsunehisa Yamamoto
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Tomoko Betsuyaku
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Ken Shinmura
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Weifeng Shen
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Eric Vivier
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Keiichi Fukuda
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| | - Motoaki Sano
- From the Department of Cardiology, Rui Jin Hospital, Medical School of Jiaotong University, Shanghai, PR China (X.Y., W.S.); Departments of Cardiology (X.Y., J.E., A.A., T.M., Y.K., K.I., T.Y., K.F., M.S.), Pulmonary Medicine (A.E.H., T.B.), and Geriatric Medicine (K.S.), Keio University School of Medicine, Tokyo, Japan; Centre d’Immunologie de Marseille-Luminy, Université de la Méditerranée, INSERM, Marseille, France (E.V.); Precursory Research for Embryonic Science and Technology (PRESTO), Japan
| |
Collapse
|
49
|
Flagellin treatment prevents increased susceptibility to systemic bacterial infection after injury by inhibiting anti-inflammatory IL-10+ IL-12- neutrophil polarization. PLoS One 2014; 9:e85623. [PMID: 24454904 PMCID: PMC3893295 DOI: 10.1371/journal.pone.0085623] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/05/2013] [Indexed: 12/28/2022] Open
Abstract
Severe trauma renders patients susceptible to infection. In sepsis, defective bacterial clearance has been linked to specific deviations in the innate immune response. We hypothesized that innate immune modulations observed during sepsis also contribute to increased bacterial susceptibility after severe trauma. A well-established murine model of burn injury, used to replicate infection following trauma, showed that wound inoculation with P. aeruginosa quickly spreads systemically. The systemic IL-10/IL-12 axis was skewed after burn injury with infection as indicated by a significant elevation in serum IL-10 and polarization of neutrophils into an anti-inflammatory ("N2"; IL-10(+) IL-12(-)) phenotype. Infection with an attenuated P. aeruginosa strain (ΔCyaB) was cleared better than the wildtype strain and was associated with an increased pro-inflammatory neutrophil ("N1"; IL-10(-)IL-12(+)) response in burn mice. This suggests that neutrophil polarization influences bacterial clearance after burn injury. Administration of a TLR5 agonist, flagellin, after burn injury restored the neutrophil response towards a N1 phenotype resulting in an increased clearance of wildtype P. aeruginosa after wound inoculation. This study details specific alterations in innate cell populations after burn injury that contribute to increased susceptibility to bacterial infection. In addition, for the first time, it identifies neutrophil polarization as a therapeutic target for the reversal of bacterial susceptibility after injury.
Collapse
|
50
|
Hall LJ, Murphy CT, Quinlan A, Hurley G, Shanahan F, Nally K, Melgar S. Natural killer cells protect mice from DSS-induced colitis by regulating neutrophil function via the NKG2A receptor. Mucosal Immunol 2013; 6:1016-26. [PMID: 23340823 DOI: 10.1038/mi.2012.140] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 12/14/2012] [Indexed: 02/04/2023]
Abstract
Natural killer (NK) cells are traditionally considered in the context of tumor surveillance and infection defense but their role in chronic inflammatory disorders such as inflammatory bowel disease is less clear. Here, we investigated the role of NK cells in dextran sodium sulfate (DSS)-induced colitis in mice. Depletion of NK cells impairs the survival of mice with colitis and is linked with dramatic increases in colonic damage, leukocyte infiltration, and pro-inflammatory profiles. Mice depleted of NK cells had increased numbers of neutrophils in colons and mesenteric lymph nodes, compared with control mice, in addition to acquiring a hyper-activation status. In vitro and in vivo studies demonstrate that NK cells downregulate pro-inflammatory functions of activated neutrophils, including reactive oxygen species and cytokine production, by direct cell-to-cell contact involving the NK cell-inhibitory receptor NKG2A. Our results indicate an immunoregulatory mechanism of action of NK cells attenuating DSS-induced colitis neutrophil-mediated inflammation and tissue injury via NKG2A-dependent mechanisms.
Collapse
Affiliation(s)
- L J Hall
- Alimentary Pharmabiotic Centre, University College Cork, National University of Ireland, Cork, Ireland.
| | | | | | | | | | | | | |
Collapse
|