1
|
Meng F, Jiang X, Wang X, Zheng Q, Wang XN, Mei C, Yan S, He Y, Xue J, Zhang X, Fu W, You Y, Zhai J, Wang Y, Sun X. Tumor necrosis factor-like cytokine 1A plays a role in inflammatory bowel disease pathogenesis. Proc Natl Acad Sci U S A 2023; 120:e2120771120. [PMID: 37579137 PMCID: PMC10452302 DOI: 10.1073/pnas.2120771120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 07/03/2023] [Indexed: 08/16/2023] Open
Abstract
The binding of tumor necrosis factor-like cytokine 1A (TL1A) to death receptor 3 (DR3) plays an important role in the interaction between dendritic cells (DCs) and T cells and contributes to intestinal inflammation development. However, the mechanism by which DCs expressing TL1A mediate helper T (Th) cell differentiation in the intestinal lamina propria (LP) during the pathogenesis of inflammatory bowel disease remains unclear. In this study, we found that TL1A/DR3 promoted Th1 and Th17 cell differentiation in T-T and DC-T cell interaction-dependent manners. TL1A-deficient CD4+ T cells failed to polarize into Th1/Th17 cells and did not cause colonic inflammation in a T cell transfer colitis model. Notably, TL1A was located in the cytoplasm and nuclei of DCs, positively regulated the DC-specific ICAM-grabbing nonintegrin/RAF1/nuclear factor κB signaling pathway, enhanced the antigen uptake ability of DCs, and promoted TLR4-mediated DC activation, inducing naive CD4+ T cell differentiation into Th1 and Th17 cells. Our work reveals that TL1A plays a regulatory role in inflammatory bowel disease pathogenesis.
Collapse
Affiliation(s)
- Fanxiang Meng
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, Liaoning Province110801, China
| | - Xuefeng Jiang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Xiao Wang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Qianqian Zheng
- Department of Pathophysiology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Xiaonan N. Wang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Chenxue Mei
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province110122, China
| | - Siqi Yan
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Yuting He
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Junxiu Xue
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| | - Xiaoqing Zhang
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Teaching Center for Medical Experiment, China Medical University, Shenyang, Liaoning Province110122, China
| | - Wenda Fu
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Blood Transfusion, Tangdu Hospital, The Fourth Military Medical University, Xi’an, Shaanxi Province710032, China
| | - Yong You
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
- Department of Immunology, Chengde medical university, Chengde, Hebei Province067000, China
| | - Jingbo Zhai
- Medical College, Inner Mongolia Minzu University, Tongliao028000, China
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Tongliao028000, China
| | - Yuanyuan Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province110032, China
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning Province110122, China
| |
Collapse
|
2
|
Liang Z, Wang Y, Lai Y, Zhang J, Yin L, Yu X, Zhou Y, Li X, Song Y. Host defense against the infection of Klebsiella pneumoniae: New strategy to kill the bacterium in the era of antibiotics? Front Cell Infect Microbiol 2022; 12:1050396. [PMID: 36506034 PMCID: PMC9730340 DOI: 10.3389/fcimb.2022.1050396] [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: 09/21/2022] [Accepted: 11/10/2022] [Indexed: 11/25/2022] Open
Abstract
Klebsiella pneumoniae (K. pneumoniae) is a typical gram-negative iatrogenic bacterium that often causes bacteremia, pneumonia and urinary tract infection particularly among those with low immunity. Although antibiotics is the cornerstone of anti-infections, the clinical efficacy of β-lactamase and carbapenems drugs has been weakened due to the emergence of drug-resistant K. pneumoniae. Recent studies have demonstrated that host defense plays a critical role in killing K. pneumoniae. Here, we summarize our current understanding of host immunity mechanisms against K. pneumoniae, including mechanical barrier, innate immune cells, cellular immunity and humoral immunity, providing a theoretical basis and the new strategy for the clinical treatment of K. pneumoniae through improving host immunity.
Collapse
Affiliation(s)
- Zihan Liang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Yiyao Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Yixiang Lai
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Jingyi Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Lanlan Yin
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Xiang Yu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Yongqin Zhou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China
| | - Xinzhi Li
- College of Basic Medical Science, China Three Gorges University, Yichang, China,Affiliated Renhe Hospital of China Three Gorges University, Yichang, China,*Correspondence: Yinhong Song, ; Xinzhi Li,
| | - Yinhong Song
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, China,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China,College of Basic Medical Science, China Three Gorges University, Yichang, China,*Correspondence: Yinhong Song, ; Xinzhi Li,
| |
Collapse
|
3
|
Roy R, Zayas J, Singh SK, Delgado K, Wood SJ, Mohamed MF, Frausto DM, Estupinian R, Giurini EF, Kuzel TM, Zloza A, Reiser J, Shafikhani SH. Overriding impaired FPR chemotaxis signaling in diabetic neutrophil stimulates infection control in murine diabetic wound. eLife 2022; 11:72071. [PMID: 35112667 PMCID: PMC8846594 DOI: 10.7554/elife.72071] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 02/01/2022] [Indexed: 11/19/2022] Open
Abstract
Infection is a major co-morbidity that contributes to impaired healing in diabetic wounds. Although impairments in diabetic neutrophils have been blamed for this co-morbidity, what causes these impairments and whether they can be overcome, remain largely unclear. Diabetic neutrophils, isolated from diabetic individuals, exhibit chemotaxis impairment but this peculiar functional impairment has been largely ignored because it appears to contradict the clinical findings which blame excessive neutrophil influx as a major impediment to healing in chronic diabetic ulcers. Here, we report that exposure to glucose in diabetic range results in impaired chemotaxis signaling through the formyl peptide receptor (FPR) in neutrophils, culminating in reduced chemotaxis and delayed neutrophil trafficking in the wound of Leprdb (db/db) type two diabetic mice, rendering diabetic wound vulnerable to infection. We further show that at least some auxiliary receptors remain functional under diabetic conditions and their engagement by the pro-inflammatory cytokine CCL3, overrides the requirement for FPR signaling and substantially improves infection control by jumpstarting the neutrophil trafficking toward infection, and stimulates healing in diabetic wound. We posit that CCL3 may have therapeutic potential for the treatment of diabetic foot ulcers if it is applied topically after the surgical debridement process which is intended to reset chronic ulcers into acute fresh wounds.
Collapse
Affiliation(s)
- Ruchi Roy
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Janet Zayas
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Sunil K Singh
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, United States
| | - Kaylee Delgado
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Stephen J Wood
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, United States
| | - Mohamed F Mohamed
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Dulce M Frausto
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Ricardo Estupinian
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Eileena F Giurini
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Timothy M Kuzel
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Andrew Zloza
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Jochen Reiser
- Department of Medicine, Rush University Medical Center, Chicago, United States
| | - Sasha H Shafikhani
- Department of Medicine, Rush University Medical Center, Chicago, United States
| |
Collapse
|
4
|
Hay AM, Howie HL, Gorham JD, D'Alessandro A, Spitalnik SL, Hudson KE, Zimring JC. Mouse background genetics in biomedical research: The devil's in the details. Transfusion 2021; 61:3017-3025. [PMID: 34480352 DOI: 10.1111/trf.16628] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Genetically modified mice are used widely to explore mechanisms in most biomedical fields-including transfusion. Concluding that a gene modification is responsible for a phenotypic change assumes no other differences between the gene-modified and wild-type mice besides the targetted gene. STUDY DESIGN AND METHODS To test the hypothesis that the N-terminus of Band3, which regulates metabolism, affects RBC storage biology, RBCs from mice with a modified N-terminus of Band3 were stored under simulated blood bank conditions. All strains of mice were generated with the same initial embryonic stem cells from 129 mice and each strain was backcrossed with C57BL/6 (B6) mice. Both 24-h recoveries post-transfusion and metabolomics were determined for stored RBCs. Genetic profiles of mice were assessed by a high-resolution SNP array. RESULTS RBCs from mice with a mutated Band3 N-terminus had increased lipid oxidation and worse 24-h recoveries, "demonstrating" that Band3 regulates oxidative injury during RBC storage. However, SNP analysis demonstrated variable inheritance of 129 genetic elements between strains. Controlled interbreeding experiments demonstrated that the changes in lipid oxidation and some of the decreased 24-hr recovery were caused by inheritance of a region of chromosome 1 of 129 origin, and not due to the modification of Band 3. SNP genotyping of a panel of commonly used commercially available KO mice showed considerable 129 contamination, despite wild-type B6 mice being listed as the correct control. DISCUSSION Thousands of articles published each year use gene-modified mice, yet genetic background issues are rarely considered. Assessment of such issues are not, but should become, routine norms of murine experimentation.
Collapse
Affiliation(s)
- Ariel M Hay
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Heather L Howie
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - James D Gorham
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Angelo D'Alessandro
- University of Colorado Denver, Anschutz Medical Campus, Denver, Colorado, USA
| | - Steven L Spitalnik
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - Krystalyn E Hudson
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, USA
| | - James C Zimring
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
5
|
Guo Y, Kasahara S, Jhingran A, Tosini NL, Zhai B, Aufiero MA, Mills KA, Gjonbalaj M, Espinosa V, Rivera A, Luster AD, Hohl TM. During Aspergillus Infection, Monocyte-Derived DCs, Neutrophils, and Plasmacytoid DCs Enhance Innate Immune Defense through CXCR3-Dependent Crosstalk. Cell Host Microbe 2020; 28:104-116.e4. [PMID: 32485165 PMCID: PMC7263227 DOI: 10.1016/j.chom.2020.05.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 01/19/2023]
Abstract
Aspergillus fumigatus, a ubiquitous mold, is a common cause of invasive aspergillosis (IA) in immunocompromised patients. Host defense against IA relies on lung-infiltrating neutrophils and monocyte-derived dendritic cells (Mo-DCs). Here, we demonstrate that plasmacytoid dendritic cells (pDCs), which are prototypically antiviral cells, participate in innate immune crosstalk underlying mucosal antifungal immunity. Aspergillus-infected murine Mo-DCs and neutrophils recruited pDCs to the lung by releasing the CXCR3 ligands, CXCL9 and CXCL10, in a Dectin-1 and Card9- and type I and III interferon signaling-dependent manner, respectively. During aspergillosis, circulating pDCs entered the lung in response to CXCR3-dependent signals. Via targeted pDC ablation, we found that pDCs were essential for host defense in the presence of normal neutrophil and Mo-DC numbers. Although interactions between pDC and fungal cells were not detected, pDCs regulated neutrophil NADPH oxidase activity and conidial killing. Thus, pDCs act as positive feedback amplifiers of neutrophil effector activity against inhaled mold conidia.
Collapse
Affiliation(s)
- Yahui Guo
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shinji Kasahara
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anupam Jhingran
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicholas L. Tosini
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bing Zhai
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mariano A. Aufiero
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kathleen A.M. Mills
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School, New York, NY, USA
| | - Mergim Gjonbalaj
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vanessa Espinosa
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | - Amariliz Rivera
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA,Department of Pediatrics, New Jersey Medical School, Rutgers Biomedical and Health Sciences (RBHS), Newark, NJ, USA
| | - Andrew D. Luster
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tobias M. Hohl
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School, New York, NY, USA,Corresponding author
| |
Collapse
|
6
|
Dabrowski AN, Conrad C, Behrendt U, Shrivastav A, Baal N, Wienhold SM, Hackstein H, N'Guessan PD, Aly S, Reppe K, Suttorp N, Zahlten J. Peptidoglycan Recognition Protein 2 Regulates Neutrophil Recruitment Into the Lungs After Streptococcus pneumoniae Infection. Front Microbiol 2019; 10:199. [PMID: 30837960 PMCID: PMC6389715 DOI: 10.3389/fmicb.2019.00199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/24/2019] [Indexed: 12/22/2022] Open
Abstract
Peptidoglycan (PGN) recognition proteins (PGLYRPs) are a highly conserved group of host defense proteins in insects and mammals that sense bacterial cell wall PGN and act bactericidally or cleave PGN by amidase function. Streptococcus (S.) pneumoniae is one of the top five killers worldwide and causes, e.g., pneumonia, endocarditis, meningitis and sepsis. S. pneumoniae accounts for approximately 1.5–2 million deaths every year. The risk of antibiotic resistance and a general poor prognosis in young children and elderly people have led to the need for new treatment approaches. To the best of our knowledge, there is no report on the relevance of PGLYRP2 in lung infections. Therefore, we infected mice deficient for PGLYRP2 transnasally with S. pneumoniae and examined the innate immune response in comparison to WT animals. As expected, PGLYRP2-KO animals had to be sacrificed earlier than their WT counterparts, and this was due to higher bacteremia. The higher bacterial load in the PGLYRP2-KO mice was accomplished with lower amounts of proinflammatory cytokines in the lungs. This led to an abolished recruitment of neutrophils into the lungs, the spread of bacteria and the subsequent aggravated course of the disease and early mortality of the PGLYRP2-KO mice. These data suggest a substantial role of PGLYRP2 in the early defense against S. pneumoniae infection, and PGLYRP2 might also affect other infections in the lungs.
Collapse
Affiliation(s)
- Alexander N Dabrowski
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Claudia Conrad
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ulrike Behrendt
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anshu Shrivastav
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nelli Baal
- Immunology and Transfusion Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Sandra M Wienhold
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Holger Hackstein
- Immunology and Transfusion Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Philippe D N'Guessan
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sahar Aly
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katrin Reppe
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Division of Pulmonary Inflammation, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Janine Zahlten
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
7
|
Baal N, Cunningham S, Obermann HL, Thomas J, Lippitsch A, Dietert K, Gruber AD, Kaufmann A, Michel G, Nist A, Stiewe T, Rupp O, Goesmann A, Zukunft S, Fleming I, Bein G, Lohmeyer J, Bauer S, Hackstein H. ADAR1 Is Required for Dendritic Cell Subset Homeostasis and Alveolar Macrophage Function. THE JOURNAL OF IMMUNOLOGY 2019; 202:1099-1111. [DOI: 10.4049/jimmunol.1800269] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 12/07/2018] [Indexed: 11/19/2022]
|
8
|
Characterization of immune cell subtypes in three commonly used mouse strains reveals gender and strain-specific variations. J Transl Med 2019; 99:93-106. [PMID: 30353130 PMCID: PMC6524955 DOI: 10.1038/s41374-018-0137-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/22/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022] Open
Abstract
The lack of consensus on bone marrow (BM) and splenic immune cell profiles in preclinical mouse strains complicates comparative analysis across different studies. Although studies have documented relative distribution of immune cells from peripheral blood in mice, similar studies for BM and spleen from naïve mice are lacking. In an effort to establish strain- and gender-specific benchmarks for distribution of various immune cell subtypes in these organs, we performed immunophenotypic analysis of BM cells and splenocytes from both genders of three commonly used murine strains (C57BL/6NCr, 129/SvHsd, and BALB/cAnNCr). Total neutrophils and splenic macrophages were significantly higher in C57BL/6NCr, whereas total B cells were lower. Within C57BL/6NCr female mice, BM B cells were elevated with respect to the males whereas splenic mDCs and splenic neutrophils were reduced. Within BALB/cAnNCr male mice, BM CD4+ Tregs were elevated with respect to the other strains. Furthermore, in male BALB/cAnNCr mice, NK cells were elevated with respect to the other strains in both BM and spleen. Splenic CD4+ Tregs and splenic CD8+ T cells were reduced in male BALB/c mice in comparison to female mice. Bone marrow CD4+ T cells and mDCs were significantly increased in 129/SvHsd whereas splenic CD8+ T cells were reduced. In general, males exhibited higher immature myeloid cells, macrophages, and NK cells. To our knowledge, this study provides a first attempt to systematically establish organ-specific benchmarks on immune cells in studies involving these mouse strains.
Collapse
|
9
|
Shrivastav A, Dabrowski AN, Conrad C, Baal N, Hackstein H, Plog S, Dietert K, Gruber AD, N'Guessan PD, Aly S, Suttorp N, Zahlten J. Peptidoglycan Recognition Protein 3 Does Not Alter the Outcome of Pneumococcal Pneumonia in Mice. Front Microbiol 2018; 9:103. [PMID: 29449834 PMCID: PMC5799233 DOI: 10.3389/fmicb.2018.00103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 01/17/2018] [Indexed: 01/07/2023] Open
Abstract
Pneumococci frequently cause community-acquired pneumonia, a disease with high mortality rates, particularly in young children and in the elderly. Endogenous antimicrobial peptides and proteins such as PGLYRP3 may contribute to the progression and outcome of this disease. Since increasing antibiotic resistant strains occur all over the world, these endogenous antimicrobial molecules are interesting new targets for future therapies. In this study, the expression pattern of PGLYRP3 was analyzed in alveolar epithelial cells, alveolar macrophages and neutrophils. Additionally, the function of PGLYRP3 during Streptococcus pneumoniae-induced pneumonia was investigated in a murine pneumococcal pneumonia model using PGLYRP3KO mice. PGLYRP3 is expressed in all selected cell types but pneumococcus-dependent induction of PGLYRP3 was observed only in neutrophils and alveolar macrophages. Interestingly, there were no significant differences in the bacterial loads within the lungs, the blood or the spleens, in the cytokine response, the composition of immune cells and the histopathology between wild type and PGLYRP3KO mice. Finally, we could neither observe significant differences in the clinical symptoms nor in the overall survival. Collectively, PGLYRP3 seems to be dispensable for the antibacterial defense during pneumococcal pneumonia.
Collapse
Affiliation(s)
- Anshu Shrivastav
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander N Dabrowski
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Conrad
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Nelli Baal
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-Universität Giessen, Giessen, Germany
| | - Stephanie Plog
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Kristina Dietert
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Philippe D N'Guessan
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sahar Aly
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Janine Zahlten
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
10
|
Sharma P, Sharma A, Srivastava M. In vivo neutralization of α4 and β7 integrins inhibits eosinophil trafficking and prevents lung injury during tropical pulmonary eosinophilia in mice. Eur J Immunol 2017; 47:1501-1512. [PMID: 28736941 DOI: 10.1002/eji.201747086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 05/27/2017] [Accepted: 07/20/2017] [Indexed: 01/21/2023]
Abstract
Integrins regulate leukocyte trafficking during homeostasis and inflammatory conditions. However, the role of α4 and β7 integrins in guiding eosinophil transmigration into the lungs during filarial manifestation of Tropical Pulmonary Eosinophilia (TPE) has not been explored. In this study, mice exhibiting TPE manifestations were administered with in vivo neutralizing antibodies against integrins α4 and β7 or their combination and immuno-pathological parameters were evaluated. Results show an intact lung barrier, significantly lower lung inflammation and reduced eosinophil counts in the Bronchoalveolar lavage fluid and lungs of mice receiving anti-α4+ β7 treatment. Reduced eosinophil peroxidase and β-hexosaminidase activity, downregulation of inflammatory genes, lower production of inflammatory lipid intermediates like prostaglandins E2 and D2, leukotriene B4 and cysteinyl leukotrienes were also noted in anti-α4+ β7 treated mice. Reduced accumulation of central memory, effector memory, regulatory T cells and lower production of IL-4, IL-5, and TGF-β were other cardinal features of anti-α4+ β7 treated mice lungs. Flow cytometry-sorted lung eosinophils from anti-α4+ β7 treated mice showed higher apoptotic potential, downregulated anti-apoptotic gene Bcl-2, and exhibited reduced F-actin polymerization and calcium influx as compared to IgG controls. In summary, neutralization of α4+ β7 integrins impairs the transmigration, activation and survival of eosinophils and reduces TPE induced pathology in mice lungs.
Collapse
Affiliation(s)
- Pankaj Sharma
- Parasitology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Aditi Sharma
- Parasitology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Mrigank Srivastava
- Parasitology Division, CSIR-Central Drug Research Institute, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| |
Collapse
|
11
|
Chakraborty K, Chatterjee S, Bhattacharyya A. Modulation of CD11c+ lung dendritic cells in respect to TGF-β in experimental pulmonary fibrosis. Cell Biol Int 2017; 41:991-1000. [PMID: 28557137 DOI: 10.1002/cbin.10800] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 05/25/2017] [Indexed: 12/30/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a deadly, progressive lung disease with very few treatment options till now. Bleomycin-induced pulmonary fibrosis (BIPF) is a commonly used mice model in IPF research. TGF-β1 has been shown to play a key role in pulmonary fibrosis (PF). Dendritic cell (DC) acts as a bridge between innate and adaptive immune systems. The coexistence of chronic inflammation sustained by mature DCs with fibrosis suggests that inflammatory phenomenon has key importance in the pathogenesis of pulmonary fibrosis. Here, we investigated the modulation of DCs phenotypic maturation, accumulation in lung tissue, and expression of other lung DC subsets in respect to TGF-β in PF. First, we established BIPF model in mice and blocked TGF-β expression by the use of inhibitor SB431542. Accumulation of lung CD11c+ DCs is significantly higher in both inflammatory and fibrotic phases of the disease but that percentages got reduced in the absence of TGF-β. TGF-β initiates up-regulation of costimulatory molecules CD86 and CD80 in the inflammatory phases of the disease but not so at fibrotic stage. Expression of lung DC subset CD11c+CD103+ is significantly increased in inflammatory phase and also in fibrotic phase of BIPF. Blocking of TGF-β causes decreased expression of CD11c+CD103+ DCs. Another important lung DC subset CD11c+CD11b+ expression is suppressed by the absence of TGF-β after bleomycin administration. CD11c+CD103+ DCs might have anti-inflammatory as well as anti-fibrotic nature in PF. All these data demonstrate differential modulation of CD11c+ lung DCs by TGF-β in experimental PF.
Collapse
Affiliation(s)
- Kaustav Chakraborty
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Soumya Chatterjee
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India
| |
Collapse
|
12
|
Karimi Y, Poznanski SM, Vahedi F, Chen B, Chew MV, Lee AJ, Ashkar AA. Type I interferon signalling is not required for the induction of endotoxin tolerance. Cytokine 2017; 95:7-11. [DOI: 10.1016/j.cyto.2017.01.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/15/2017] [Accepted: 01/25/2017] [Indexed: 11/28/2022]
|
13
|
Lippitsch A, Chukovetskyi Y, Baal N, Bein G, Hackstein H. Unique high and homogenous surface expression of the transferrin receptor CD71 on murine plasmacytoid dendritic cells in different tissues. Cell Immunol 2017; 316:41-52. [PMID: 28372797 DOI: 10.1016/j.cellimm.2017.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 02/28/2017] [Accepted: 03/27/2017] [Indexed: 01/03/2023]
Abstract
Plasmacytoid dendritic cells (pDC) are of increasing interest in cancer vaccine development, but many functions of these highly specialized, multifaceted cells are poorly understood. The transferrin receptor CD71 has also been suggested to function as an antigen uptake receptor on professional antigen-presenting cells. In this study, we employed multiparameter flow cytometry to investigate CD71 expression on various leukocyte subsets, including DC subsets, granulocytes, macrophages, T and B lymphocytes, γδ T cells, and natural killer cells. Cells from various lymphoid and non-lymphoid murine tissues were analyzed using fluorochrome-conjugated monoclonal antibodies. High CD71 expression (90-100%) was observed, uniquely on pDC amongst the leukocyte populations examined, in both lymphoid and non-lymphoid tissues, including other DC subsets. In contrast, CD71 expression on non-tissue pDC, in the bone marrow and peripheral blood, was reduced. The cause and function of this high tissue pDC-selective CD71 expression remain to be examined.
Collapse
Affiliation(s)
- Anne Lippitsch
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany
| | - Yuri Chukovetskyi
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany
| | - Nelli Baal
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany
| | - Gregor Bein
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany
| | - Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Biomedical Research Center Seltersberg (BFS), Schubertstrasse 81, D-35392 Giessen, Germany.
| |
Collapse
|
14
|
Beauchemin KJ, Wells JM, Kho AT, Philip VM, Kamir D, Kohane IS, Graber JH, Bult CJ. Temporal dynamics of the developing lung transcriptome in three common inbred strains of laboratory mice reveals multiple stages of postnatal alveolar development. PeerJ 2016; 4:e2318. [PMID: 27602285 PMCID: PMC4991849 DOI: 10.7717/peerj.2318] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/12/2016] [Indexed: 12/12/2022] Open
Abstract
To characterize temporal patterns of transcriptional activity during normal lung development, we generated genome wide gene expression data for 26 pre- and post-natal time points in three common inbred strains of laboratory mice (C57BL/6J, A/J, and C3H/HeJ). Using Principal Component Analysis and least squares regression modeling, we identified both strain-independent and strain-dependent patterns of gene expression. The 4,683 genes contributing to the strain-independent expression patterns were used to define a murine Developing Lung Characteristic Subtranscriptome (mDLCS). Regression modeling of the Principal Components supported the four canonical stages of mammalian embryonic lung development (embryonic, pseudoglandular, canalicular, saccular) defined previously by morphology and histology. For postnatal alveolar development, the regression model was consistent with four stages of alveolarization characterized by episodic transcriptional activity of genes related to pulmonary vascularization. Genes expressed in a strain-dependent manner were enriched for annotations related to neurogenesis, extracellular matrix organization, and Wnt signaling. Finally, a comparison of mouse and human transcriptomics from pre-natal stages of lung development revealed conservation of pathways associated with cell cycle, axon guidance, immune function, and metabolism as well as organism-specific expression of genes associated with extracellular matrix organization and protein modification. The mouse lung development transcriptome data generated for this study serves as a unique reference set to identify genes and pathways essential for normal mammalian lung development and for investigations into the developmental origins of respiratory disease and cancer. The gene expression data are available from the Gene Expression Omnibus (GEO) archive (GSE74243). Temporal expression patterns of mouse genes can be investigated using a study specific web resource (http://lungdevelopment.jax.org).
Collapse
Affiliation(s)
- Kyle J. Beauchemin
- The Jackson Laboratory, Bar Harbor, ME, United States
- Graduate School of Biomedical Sciences and Engineering, The University of Maine, Orono, ME, United States
| | | | - Alvin T. Kho
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA, United States
| | | | - Daniela Kamir
- The Jackson Laboratory, Bar Harbor, ME, United States
| | - Isaac S. Kohane
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, United States
| | | | - Carol J. Bult
- The Jackson Laboratory, Bar Harbor, ME, United States
| |
Collapse
|
15
|
Stolt C, Schmidt IHE, Sayfart Y, Steinmetz I, Bast A. Heme Oxygenase-1 and Carbon Monoxide PromoteBurkholderia pseudomalleiInfection. THE JOURNAL OF IMMUNOLOGY 2016; 197:834-46. [DOI: 10.4049/jimmunol.1403104] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 05/26/2016] [Indexed: 12/25/2022]
|
16
|
Hoffmann F, Ender F, Schmudde I, Lewkowich IP, Köhl J, König P, Laumonnier Y. Origin, Localization, and Immunoregulatory Properties of Pulmonary Phagocytes in Allergic Asthma. Front Immunol 2016; 7:107. [PMID: 27047494 PMCID: PMC4803735 DOI: 10.3389/fimmu.2016.00107] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/08/2016] [Indexed: 01/21/2023] Open
Abstract
Allergic asthma is a chronic inflammatory disease of the airways that is driven by maladaptive T helper 2 (Th2) and Th17 immune responses against harmless, airborne substances. Pulmonary phagocytes represent the first line of defense in the lung where they constantly sense the local environment for potential threats. They comprise two distinct cell types, i.e., macrophages and dendritic cells (DC) that differ in their origins and functions. Alveolar macrophages quickly take up most of the inhaled allergens, yet do not deliver their cargo to naive T cells sampling in draining lymph nodes. In contrast, pulmonary DCs instruct CD4(+) T cells develop into Th2 and Th17 effectors, initiating the maladaptive immune responses toward harmless environmental substances observed in allergic individuals. Unraveling the mechanisms underlying this mistaken identity of harmless, airborne substances by innate immune cells is one of the great challenges in asthma research. The identification of different pulmonary DC subsets, their role in antigen uptake, migration to the draining lymph nodes, and their potential to instruct distinct T cell responses has set the stage to unravel this mystery. However, at this point, a detailed understanding of the spatiotemporal resolution of DC subset localization, allergen uptake, processing, autocrine and paracrine cellular crosstalk, and the humoral factors that define the activation status of DCs is still lacking. In addition to DCs, at least two distinct macrophage populations have been identified in the lung that are either located in the airway/alveolar lumen or in the interstitium. Recent data suggest that such populations can exert either pro- or anti-inflammatory functions. Similar to the DC subsets, detailed insights into the individual roles of alveolar and interstitial macrophages during the different phases of asthma development are still missing. Here, we will provide an update on the current understanding of the origin, localization, and function of the diverse pulmonary antigen-presenting cell subsets, in particular with regard to the development and regulation of allergic asthma. While most data are from mouse models of experimental asthma, we have also included available human data to judge the translational value of the findings obtained in experimental asthma models.
Collapse
Affiliation(s)
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Inken Schmudde
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ian P. Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Peter König
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
17
|
Sharma P, Sharma A, Vishwakarma AL, Agnihotri PK, Sharma S, Srivastava M. Host lung immunity is severely compromised during tropical pulmonary eosinophilia: role of lung eosinophils and macrophages. J Leukoc Biol 2015; 99:619-28. [PMID: 26489428 DOI: 10.1189/jlb.4a0715-309rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/01/2015] [Indexed: 12/28/2022] Open
Abstract
Eosinophils play a central role in the pathogenesis of tropical pulmonary eosinophilia, a rare, but fatal, manifestation of filariasis. However, no exhaustive study has been done to identify the genes and proteins of eosinophils involved in the pathogenesis of tropical pulmonary eosinophilia. In the present study, we established a mouse model of tropical pulmonary eosinophilia that mimicked filarial manifestations of human tropical pulmonary eosinophilia pathogenesis and used flow cytometry-assisted cell sorting and real-time RT-PCR to study the gene expression profile of flow-sorted, lung eosinophils and lung macrophages during tropical pulmonary eosinophilia pathogenesis. Our results show that tropical pulmonary eosinophilia mice exhibited increased levels of IL-4, IL-5, CCL5, and CCL11 in the bronchoalveolar lavage fluid and lung parenchyma along with elevated titers of IgE and IgG subtypes in the serum. Alveolar macrophages from tropical pulmonary eosinophilia mice displayed decreased phagocytosis, attenuated nitric oxide production, and reduced T-cell proliferation capacity, and FACS-sorted lung eosinophils from tropical pulmonary eosinophilia mice upregulated transcript levels of ficolin A and anti-apoptotic gene Bcl2,but proapoptotic genes Bim and Bax were downregulated. Similarly, flow-sorted lung macrophages upregulated transcript levels of TLR-2, TLR-6, arginase-1, Ym-1, and FIZZ-1 but downregulated nitric oxide synthase-2 levels, signifying their alternative activation. Taken together, we show that the pathogenesis of tropical pulmonary eosinophilia is marked by functional impairment of alveolar macrophages, alternative activation of lung macrophages, and upregulation of anti-apoptotic genes by eosinophils. These events combine together to cause severe lung inflammation and compromised lung immunity. Therapeutic interventions that can boost host immune response in the lungs might thus provide relief to patients with tropical pulmonary eosinophilia.
Collapse
Affiliation(s)
- Pankaj Sharma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Aditi Sharma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Achchhe Lal Vishwakarma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Promod Kumar Agnihotri
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Sharad Sharma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Mrigank Srivastava
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| |
Collapse
|
18
|
Hackstein H, Lippitsch A, Krug P, Schevtschenko I, Kranz S, Hecker M, Dietert K, Gruber AD, Bein G, Brendel C, Baal N. Prospectively defined murine mesenchymal stem cells inhibit Klebsiella pneumoniae-induced acute lung injury and improve pneumonia survival. Respir Res 2015; 16:123. [PMID: 26438075 PMCID: PMC4594670 DOI: 10.1186/s12931-015-0288-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 10/02/2015] [Indexed: 12/12/2022] Open
Abstract
Background Numerous studies have described the immunosuppressive capacity of mesenchymal stem cells (MSC) but these studies use mixtures of heterogeneous progenitor cells for in vitro expansion. Recently, multipotent MSC have been prospectively identified in murine bone marrow (BM) on the basis of PDFGRa+ SCA1+ CD45− TER119− (PαS) expression but the immunomodulatory capacity of these MSC is unknown. Methods We isolated PαS MSC by high-purity FACS sorting of murine BM and after in vitro expansion we analyzed the in vivo immunomodulatory activity during acute pneumonia. PαS MSC (1 × 106) were applied intratracheally 4 h after acute respiratory Klebsiella pneumoniae induced infection. Results PαS MSC treatment resulted in significantly reduced alveolitis and protein leakage in comparison to mock-treated controls. PαS MSC-treated mice exhibited significantly reduced alveolar TNF-α and IL-12p70 expression, while IL-10 expression was unaffected. Dissection of respiratory dendritic cell (DC) subsets by multiparameter flow cytometry revealed significantly reduced lung DC infiltration and significantly reduced CD86 costimulatory expression on lung CD103+ DC in PαS MSC-treated mice. In the post-acute phase of pneumonia, PαS MSC-treated animals exhibited significantly reduced respiratory IL-17+ CD4+ T cells and IFN-γ+ CD4+ T cells. Moreover, PαS MSC treatment significantly improved overall pneumonia survival and did not increase bacterial load. Conclusion In this study we demonstrated for the first time the feasibility and in vivo immunomodulatory capacity of prospectively defined MSC in pneumonia. Electronic supplementary material The online version of this article (doi:10.1186/s12931-015-0288-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Langhansstr. 7, D-35390, Giessen, Germany.
| | - Anne Lippitsch
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Langhansstr. 7, D-35390, Giessen, Germany.
| | - Philipp Krug
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Langhansstr. 7, D-35390, Giessen, Germany.
| | - Inna Schevtschenko
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Langhansstr. 7, D-35390, Giessen, Germany.
| | - Sabine Kranz
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Langhansstr. 7, D-35390, Giessen, Germany.
| | - Matthias Hecker
- Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Giessen, Germany.
| | - Kristina Dietert
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, 14163, Berlin, Germany.
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, 14163, Berlin, Germany.
| | - Gregor Bein
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Langhansstr. 7, D-35390, Giessen, Germany.
| | - Cornelia Brendel
- Department of Hematology, Oncology and Immunology, Philipps University Marburg, University Hospital Giessen und Marburg, Marburg, Germany.
| | - Nelli Baal
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Langhansstr. 7, D-35390, Giessen, Germany.
| |
Collapse
|
19
|
Pachynski RK, Scholz A, Monnier J, Butcher EC, Zabel BA. Evaluation of Tumor-infiltrating Leukocyte Subsets in a Subcutaneous Tumor Model. J Vis Exp 2015:52657. [PMID: 25938949 PMCID: PMC4541547 DOI: 10.3791/52657] [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: 01/04/2023] Open
Abstract
Specialized immune cells that infiltrate the tumor microenvironment regulate the growth and survival of neoplasia. Malignant cells must elude or subvert anti-tumor immune responses in order to survive and flourish. Tumors take advantage of a number of different mechanisms of immune "escape," including the recruitment of tolerogenic DC, immunosuppressive regulatory T cells (Tregs), and myeloid-derived suppressor cells (MDSC) that inhibit cytotoxic anti-tumor responses. Conversely, anti-tumor effector immune cells can slow the growth and expansion of malignancies: immunostimulatory dendritic cells, natural killer cells which harbor innate anti-tumor immunity, and cytotoxic T cells all can participate in tumor suppression. The balance between pro- and anti-tumor leukocytes ultimately determines the behavior and fate of transformed cells; a multitude of human clinical studies have borne this out. Thus, detailed analysis of leukocyte subsets within the tumor microenvironment has become increasingly important. Here, we describe a method for analyzing infiltrating leukocyte subsets present in the tumor microenvironment in a mouse tumor model. Mouse B16 melanoma tumor cells were inoculated subcutaneously in C57BL/6 mice. At a specified time, tumors and surrounding skin were resected en bloc and processed into single cell suspensions, which were then stained for multi-color flow cytometry. Using a variety of leukocyte subset markers, we were able to compare the relative percentages of infiltrating leukocyte subsets between control and chemerin-expressing tumors. Investigators may use such a tool to study the immune presence in the tumor microenvironment and when combined with traditional caliper size measurements of tumor growth, will potentially allow them to elucidate the impact of changes in immune composition on tumor growth. Such a technique can be applied to any tumor model in which the tumor and its microenvironment can be resected and processed.
Collapse
MESH Headings
- Animals
- Disease Models, Animal
- Female
- Flow Cytometry
- Killer Cells, Natural/immunology
- Leukocytes/immunology
- Leukocytes/pathology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Tumor Microenvironment
Collapse
Affiliation(s)
- Russell K Pachynski
- Division of Oncology, Department of Medicine, Washington University School of Medicine;
| | - Alexander Scholz
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine
| | - Justin Monnier
- Palo Alto Institute for Research and Education, Veterans Affairs Palo Alto Health Care System; Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine
| | - Brian A Zabel
- Palo Alto Institute for Research and Education, Veterans Affairs Palo Alto Health Care System
| |
Collapse
|
20
|
Oliveira THVD, Campos KKD, Soares NP, Pena KB, Lima WG, Bezerra FS. Influence of Sexual Dimorphism on Pulmonary Inflammatory Response in Adult Mice Exposed to Chloroform. Int J Toxicol 2015; 34:250-7. [DOI: 10.1177/1091581815580172] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Chloroform is an organic solvent used as an intermediate in the synthesis of various fluorocarbons. Despite its widespread use in industry and agriculture, exposure to chloroform can cause illnesses such as cancer, especially in the liver and kidneys. The aim of the study was to analyze the effects of chloroform on redox imbalance and pulmonary inflammatory response in adult C57BL/6 mice. Forty animals were divided into 4 groups (N = 10): female (FCG) and male (MCG) controls, and females (FEG) and males (MEG) exposed to chloroform (7.0 ppm) 3 times/d for 20 minutes for 5 days. Total and differential cell counts, oxidative damage analysis, and protein carbonyl and antioxidant enzyme catalase (CAT) activity measurements were performed. Morphometric analyses included alveolar area (Aa) and volume density of alveolar septa (Vv) measurements. Compared to FCG and MCG, inflammatory cell influx, oxidative damage to lipids and proteins, and CAT activity were higher in FEG and MEG, respectively. Oxidative damage and enzyme CAT activity were higher in FEG than in FCG. The Aa was higher in FEG and MEG than in FCG and MCG, respectively. The Vv was lower in FEG and MEG than in FCG and MCG, respectively. This study highlights the risks of occupational chloroform exposure at low concentrations and the intensity of oxidative damage related to gender. The results validate a model of acute exposure that provides cellular and biochemical data through short-term exposure to chloroform.
Collapse
Affiliation(s)
| | - Keila Karine Duarte Campos
- Department of Biological Sciences (DECBI), Laboratory of Metabolic Biochemistry (LBM), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Nícia Pedreira Soares
- Department of Biological Sciences (DECBI), Laboratory of Metabolic Biochemistry (LBM), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Karina Braga Pena
- Department of Biological Sciences (DECBI), Laboratory of Metabolic Biochemistry (LBM), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Wanderson Geraldo Lima
- Department of Biological Sciences (DECBI), Laboratory of Morphopathology (LMP), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| | - Frank Silva Bezerra
- Department of Biological Sciences (DECBI), Laboratory of Metabolic Biochemistry (LBM), Center of Research in Biological Sciences (NUPEB), Federal University of Ouro Preto (UFOP), Ouro Preto, Minas Gerais, Brazil
| |
Collapse
|
21
|
Paun A, Kunwar A, Haston CK. Acute adaptive immune response correlates with late radiation-induced pulmonary fibrosis in mice. Radiat Oncol 2015; 10:45. [PMID: 25889053 PMCID: PMC4342202 DOI: 10.1186/s13014-015-0359-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 02/16/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The lung response to radiation exposure can involve an immediate or early reaction to the radiation challenge, including cell death and an initial immune reaction, and can be followed by a tissue injury response, of pneumonitis or fibrosis, to this acute reaction. Herein, we aimed to determine whether markers of the initial immune response, measured within days of radiation exposure, are correlated with the lung tissue injury responses occurring weeks later. METHODS Inbred strains of mice known to be susceptible (KK/HIJ, C57BL/6J, 129S1/SvImJ) or resistant (C3H/HeJ, A/J, AKR/J) to radiation-induced pulmonary fibrosis and to vary in time to onset of respiratory distress post thoracic irradiation (from 10-23 weeks) were studied. Mice were untreated (controls) or received 18 Gy whole thorax irradiation and were euthanized at 6 h, 1d or 7 d after radiation treatment. Pulmonary CD4+ lymphocytes, bronchoalveolar cell profile & cytokine level, and serum cytokine levels were assayed. RESULTS Thoracic irradiation and inbred strain background significantly affected the numbers of CD4+ cells in the lungs and the bronchoalveolar lavage cell differential of exposed mice. At the 7 day timepoint greater numbers of pulmonary Th1 and Th17 lymphocytes and reduced lavage interleukin17 and interferonγ levels were significant predictors of late stage fibrosis. Lavage levels of interleukin-10, measured at the 7 day timepoint, were inversely correlated with fibrosis score (R=-0.80, p=0.05), while serum levels of interleukin-17 in control mice significantly correlated with post irradiation survival time (R=0.81, p=0.04). Lavage macrophage, lymphocyte or neutrophil counts were not significantly correlated with either of fibrosis score or time to respiratory distress in the six mouse strains. CONCLUSION Specific cytokine and lymphocyte levels, but not strain dependent lavage cell profiles, were predictive of later radiation-induced lung injury in this panel of inbred strains.
Collapse
Affiliation(s)
- Alexandra Paun
- Department of Human Genetics, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.
| | - Amit Kunwar
- Department of Human Genetics, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.
| | - Christina K Haston
- Department of Human Genetics, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada. .,Department of Medicine, Meakins-Christie Laboratories, McGill University, 3626 St. Urbain, H2X 2P2, Montreal, QC, Canada.
| |
Collapse
|
22
|
Alpaerts K, Buckinx R, Adriaensen D, Van Nassauw L, Timmermans JP. Identification and Putative Roles of Distinct Subtypes of Intestinal Dendritic Cells in Neuroimmune Communication: What can be Learned from Other Organ Systems? Anat Rec (Hoboken) 2015; 298:903-16. [DOI: 10.1002/ar.23106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 09/13/2014] [Accepted: 11/08/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Katrien Alpaerts
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Roeland Buckinx
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Dirk Adriaensen
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Luc Van Nassauw
- Laboratory of Human Anatomy and Embryology; Faculty of Medicine and Health Sciences; University of Antwerp; Antwerp Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| |
Collapse
|
23
|
Misharin AV, Morales-Nebreda L, Mutlu GM, Budinger GRS, Perlman H. Flow cytometric analysis of macrophages and dendritic cell subsets in the mouse lung. Am J Respir Cell Mol Biol 2014; 49:503-10. [PMID: 23672262 DOI: 10.1165/rcmb.2013-0086ma] [Citation(s) in RCA: 635] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The lung hosts multiple populations of macrophages and dendritic cells, which play a crucial role in lung pathology. The accurate identification and enumeration of these subsets are essential for understanding their role in lung pathology. Flow cytometry is a mainstream tool for studying the immune system. However, a systematic flow cytometric approach to identify subsets of macrophages and dendritic cells (DCs) accurately and consistently in the normal mouse lung has not been described. Here we developed a panel of surface markers and an analysis strategy that accurately identify all known populations of macrophages and DCs, and their precursors in the lung during steady-state conditions and bleomycin-induced injury. Using this panel, we assessed the polarization of lung macrophages during the course of bleomycin-induced lung injury. Alveolar macrophages expressed markers of alternatively activated macrophages during both acute and fibrotic phases of bleomycin-induced lung injury, whereas markers of classically activated macrophages were expressed only during the acute phase. Taken together, these data suggest that this flow cytometric panel is very helpful in identifying macrophage and DC populations and their state of activation in normal, injured, and fibrotic lungs.
Collapse
|
24
|
Bast A, Krause K, Schmidt IHE, Pudla M, Brakopp S, Hopf V, Breitbach K, Steinmetz I. Caspase-1-dependent and -independent cell death pathways in Burkholderia pseudomallei infection of macrophages. PLoS Pathog 2014; 10:e1003986. [PMID: 24626296 PMCID: PMC3953413 DOI: 10.1371/journal.ppat.1003986] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 01/25/2014] [Indexed: 12/19/2022] Open
Abstract
The cytosolic pathogen Burkholderia pseudomallei and causative agent of melioidosis has been shown to regulate IL-1β and IL-18 production through NOD-like receptor NLRP3 and pyroptosis via NLRC4. Downstream signalling pathways of those receptors and other cell death mechanisms induced during B. pseudomallei infection have not been addressed so far in detail. Furthermore, the role of B. pseudomallei factors in inflammasome activation is still ill defined. In the present study we show that caspase-1 processing and pyroptosis is exclusively dependent on NLRC4, but not on NLRP3 in the early phase of macrophage infection, whereas at later time points caspase-1 activation and cell death is NLRC4- independent. In the early phase we identified an activation pathway involving caspases-9, -7 and PARP downstream of NLRC4 and caspase-1. Analyses of caspase-1/11-deficient infected macrophages revealed a strong induction of apoptosis, which is dependent on activation of apoptotic initiator and effector caspases. The early activation pathway of caspase-1 in macrophages was markedly reduced or completely abolished after infection with a B. pseudomallei flagellin FliC or a T3SS3 BsaU mutant. Studies using cells transfected with the wild-type and mutated T3SS3 effector protein BopE indicated also a role of this protein in caspase-1 processing. A T3SS3 inner rod protein BsaK mutant failed to activate caspase-1, revealed higher intracellular counts, reduced cell death and IL-1β secretion during early but not during late macrophage infection compared to the wild-type. Intranasal infection of BALB/c mice with the BsaK mutant displayed a strongly decreased mortality, lower bacterial loads in organs, and reduced levels of IL-1β, myeloperoxidase and neutrophils in bronchoalveolar lavage fluid. In conclusion, our results indicate a major role for a functional T3SS3 in early NLRC4-mediated caspase-1 activation and pyroptosis and a contribution of late caspase-1-dependent and -independent cell death mechanisms in the pathogenesis of B. pseudomallei infection. Inflammasome activation is important for host defence against bacterial infection. Many gram-negative pathogens use secretion systems to inject bacterial proteins such as flagellin or structural components of the secretion machinery itself into the host cytosol leading to caspase-1 activation and pyroptotic cell death. However, little is known about the B. pseudomallei factors that trigger caspase-1 activation as well as downstream signalling pathways and effector mechanisms of caspase-1. Here, we identified the B. pseudomallei T3SS3 inner rod protein BsaK as an early activator of caspase-1-dependent cell death and IL-1β secretion in primary macrophages and as a virulence factor in murine melioidosis. We could show that upon infection of macrophages, caspase-7 is activated downstream of the NLRC4/caspase-1 inflammasome and requires caspase-9 processing. Although caspase-7 was essential for cleavage of the DNA damage sensor PARP during pyroptosis, it did neither contribute to cytokine production nor B. pseudomallei growth restriction by promoting early macrophage death. In addition to a rapid NLRC4/caspase-1- dependent induction of pyroptosis in wild-type macrophages, we observed a delayed activation of classical apoptosis in macrophages lacking caspase-1/11. Thus, initiation of different cell death pathways seems to be an effective strategy to limit intracellular B. pseudomallei infection.
Collapse
Affiliation(s)
- Antje Bast
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Kathrin Krause
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Imke H. E. Schmidt
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Matsayapan Pudla
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Stefanie Brakopp
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Verena Hopf
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Katrin Breitbach
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Ivo Steinmetz
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
- * E-mail:
| |
Collapse
|
25
|
Wood S, Jayaraman V, Huelsmann EJ, Bonish B, Burgad D, Sivaramakrishnan G, Qin S, DiPietro LA, Zloza A, Zhang C, Shafikhani SH. Pro-inflammatory chemokine CCL2 (MCP-1) promotes healing in diabetic wounds by restoring the macrophage response. PLoS One 2014; 9:e91574. [PMID: 24618995 PMCID: PMC3950222 DOI: 10.1371/journal.pone.0091574] [Citation(s) in RCA: 173] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 02/13/2014] [Indexed: 01/13/2023] Open
Abstract
Prior studies suggest that the impaired healing seen in diabetic wounds derives from a state of persistent hyper-inflammation characterized by harmful increases in inflammatory leukocytes including macrophages. However, such studies have focused on wounds at later time points (day 10 or older), and very little attention has been given to the dynamics of macrophage responses in diabetic wounds early after injury. Given the importance of macrophages for the process of healing, we studied the dynamics of macrophage response during early and late phases of healing in diabetic wounds. Here, we report that early after injury, the diabetic wound exhibits a significant delay in macrophage infiltration. The delay in the macrophage response in diabetic wounds results from reduced Chemokine (C-C motif) ligand 2 (CCL2) expression. Importantly, one-time treatment with chemoattractant CCL2 significantly stimulated healing in diabetic wounds by restoring the macrophage response. Our data demonstrate that, rather than a hyper-inflammatory state; the early diabetic wound exhibits a paradoxical and damaging decrease in essential macrophage response. Our studies suggest that the restoration of the proper kinetics of macrophage response may be able to jumpstart subsequent healing stages. CCL2 chemokine-based therapy may be an attractive strategy to promote healing in diabetic wounds.
Collapse
Affiliation(s)
- Stephen Wood
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Vijayakumar Jayaraman
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Erica J. Huelsmann
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
- Rush University Cancer Center, Rush University Medical Center, Chicago, Illinois, United States of America
- Developmental Center for AIDS Research, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Brian Bonish
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
- Department of Dermatology, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Derick Burgad
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Gayathri Sivaramakrishnan
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Shanshan Qin
- Department of Pharmacology, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Luisa A. DiPietro
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Andrew Zloza
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
- Rush University Cancer Center, Rush University Medical Center, Chicago, Illinois, United States of America
- Developmental Center for AIDS Research, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Chunxiang Zhang
- Department of Pharmacology, Rush University Medical Center, Chicago, Illinois, United States of America
| | - Sasha H. Shafikhani
- Department of Immunology/Microbiology, Rush University Medical Center, Chicago, Illinois, United States of America
- Rush University Cancer Center, Rush University Medical Center, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
26
|
Hackstein H, Kranz S, Lippitsch A, Wachtendorf A, Kershaw O, Gruber AD, Michel G, Lohmeyer J, Bein G, Baal N, Herold S. Modulation of respiratory dendritic cells during Klebsiella pneumonia infection. Respir Res 2013; 14:91. [PMID: 24044871 PMCID: PMC3848864 DOI: 10.1186/1465-9921-14-91] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 09/13/2013] [Indexed: 12/21/2022] Open
Abstract
Background Klebsiella pneumoniae is a leading cause of severe hospital-acquired respiratory tract infections and death but little is known regarding the modulation of respiratory dendritic cell (DC) subsets. Plasmacytoid DC (pDC) are specialized type 1 interferon producing cells and considered to be classical mediators of antiviral immunity. Method By using multiparameter flow cytometry analysis we have analysed the modulation of respiratory DC subsets after intratracheal Klebsiella pneumonia infection. Results Data indicate that pDCs and MoDC were markedly elevated in the post acute pneumonia phase when compared to mock-infected controls. Analysis of draining mediastinal lymph nodes revealed a rapid increase of activated CD103+ DC, CD11b+ DC and MoDC within 48 h post infection. Lung pDC identification during bacterial pneumonia was confirmed by extended phenotyping for 120G8, mPDCA-1 and Siglec-H expression and by demonstration of high Interferon-alpha producing capacity after cell sorting. Cytokine expression analysis of ex vivo-sorted respiratory DC subpopulations from infected animals revealed elevated Interferon-alpha in pDC, elevated IFN-gamma, IL-4 and IL-13 in CD103+ DC and IL-19 and IL-12p35 in CD11b+ DC subsets in comparison to CD11c+ MHC-class IIlow cells indicating distinct functional roles. Antigen-specific naive CD4+ T cell stimulatory capacity of purified respiratory DC subsets was analysed in a model system with purified ovalbumin T cell receptor transgenic naive CD4+ responder T cells and respiratory DC subsets, pulsed with ovalbumin and matured with Klebsiella pneumoniae lysate. CD103+ DC and CD11b+ DC subsets represented the most potent naive CD4+ T helper cell activators. Conclusion These results provide novel insight into the activation of respiratory DC subsets during Klebsiella pneumonia infection. The detection of increased respiratory pDC numbers in bacterial pneumonia may indicate possible novel pDC functions with respect to lung repair and regeneration.
Collapse
Affiliation(s)
- Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Member of the German Center for Lung Research (DZL), Langhansstr, 7, D-35392, Giessen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Hackstein H, Hagel N, Knoche A, Kranz S, Lohmeyer J, von Wulffen W, Kershaw O, Gruber AD, Bein G, Baal N. Skin TLR7 triggering promotes accumulation of respiratory dendritic cells and natural killer cells. PLoS One 2012; 7:e43320. [PMID: 22927956 PMCID: PMC3425551 DOI: 10.1371/journal.pone.0043320] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 07/19/2012] [Indexed: 01/08/2023] Open
Abstract
The TLR7 agonist imiquimod has been used successfully as adjuvant for skin treatment of virus-associated warts and basal cell carcinoma. The effects of skin TLR7 triggering on respiratory leukocyte populations are unknown. In a placebo-controlled experimental animal study we have used multicolour flow cytometry to systematically analyze the modulation of respiratory leukocyte subsets after skin administration of imiquimod. Compared to placebo, skin administration of imiquimod significantly increased respiratory dendritic cells (DC) and natural killer cells, whereas total respiratory leukocyte, alveolar macrophages, classical CD4+ T helper and CD8+ T killer cell numbers were not or only moderately affected. DC subpopulation analyses revealed that elevation of respiratory DC was caused by an increase of respiratory monocytic DC and CD11bhi DC subsets. Lymphocyte subpopulation analyses indicated a marked elevation of respiratory natural killer cells and a significant reduction of B lymphocytes. Analysis of cytokine responses of respiratory leukocytes after stimulation with Klebsiella pneumonia indicated reduced IFN-γ and TNF-α expression and increased IL-10 and IL-12p70 production after 7 day low dose skin TLR7 triggering. Additionally, respiratory NK cytotoxic activity was increased after 7d skin TLR7 triggering. In contrast, lung histology and bronchoalveolar cell counts were not affected suggesting that skin TLR7 stimulation modulated respiratory leukocyte composition without inducing overt pulmonary inflammation. These data suggest the possibility to modulate respiratory leukocyte composition and respiratory cytokine responses against pathogens like Klebsiella pneumonia through skin administration of a clinically approved TLR7 ligand. Skin administration of synthetic TLR7 ligands may represent a novel, noninvasive means to modulate respiratory immunity.
Collapse
Affiliation(s)
- Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Justus-Liebig-University Giessen, Member of the German Center for Lung Research, Giessen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|