1
|
Zhang Y, Zhang Z, Mo Y, Zhang Y, Yuan J, Zhang Q. MMP-3 mediates copper oxide nanoparticle-induced pulmonary inflammation and fibrosis. J Nanobiotechnology 2024; 22:428. [PMID: 39030581 PMCID: PMC11264740 DOI: 10.1186/s12951-024-02707-x] [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: 05/02/2024] [Accepted: 07/05/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND The increasing production and usage of copper oxide nanoparticles (Nano-CuO) raise human health concerns. Previous studies have demonstrated that exposure to Nano-CuO could induce lung inflammation, injury, and fibrosis. However, the potential underlying mechanisms are still unclear. Here, we proposed that matrix metalloproteinase-3 (MMP-3) might play an important role in Nano-CuO-induced lung inflammation, injury, and fibrosis. RESULTS Exposure of mice to Nano-CuO caused acute lung inflammation and injury in a dose-dependent manner, which was reflected by increased total cell number, neutrophil count, macrophage count, lactate dehydrogenase (LDH) activity, and CXCL1/KC level in bronchoalveolar lavage fluid (BALF) obtained on day 3 post-exposure. The time-response study showed that Nano-CuO-induced acute lung inflammation and injury appeared as early as day 1 after exposure, peaked on day 3, and ameliorated over time. However, even on day 42 post-exposure, the LDH activity and macrophage count were still higher than those in the control group, suggesting that Nano-CuO caused chronic lung inflammation. The Nano-CuO-induced pulmonary inflammation was further confirmed by H&E staining of lung sections. Trichrome staining showed that Nano-CuO exposure caused pulmonary fibrosis from day 14 to day 42 post-exposure with an increasing tendency over time. Increased hydroxyproline content and expression levels of fibrosis-associated proteins in mouse lungs were also observed. In addition, Nano-CuO exposure induced MMP-3 overexpression and increased MMP-3 secretion in mouse lungs. Knocking down MMP-3 in mouse lungs significantly attenuated Nano-CuO-induced acute and chronic lung inflammation and fibrosis. Moreover, Nano-CuO exposure caused sustained production of cleaved osteopontin (OPN) in mouse lungs, which was also significantly decreased by knocking down MMP-3. CONCLUSIONS Our results demonstrated that short-term Nano-CuO exposure caused acute lung inflammation and injury, while long-term exposure induced chronic pulmonary inflammation and fibrosis. Knocking down MMP-3 significantly ameliorated Nano-CuO-induced pulmonary inflammation, injury, and fibrosis, and also attenuated Nano-CuO-induced cleaved OPN level. Our study suggests that MMP-3 may play important roles in Nano-CuO-induced pulmonary inflammation and fibrosis via cleavage of OPN and may provide a further understanding of the mechanisms underlying Nano-CuO-induced pulmonary toxicity.
Collapse
Affiliation(s)
- Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
- Beijing Key Laboratory of Occupational Safety and Health, Institute of Urban Safety and Environmental Science, Beijing Academy of Science and Technology, Beijing, 100054, China
| | - Zhenyu Zhang
- Department of Emergency, Xiang'An Hospital of Xiamen University, Xiamen, 361104, Fujian, China
| | - Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Yue Zhang
- Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, 485 E. Gray Street, Louisville, KY, 40202, USA.
| |
Collapse
|
2
|
Hoffmann JP, Srivastava A, Yang H, Iwanaga N, Remcho TP, Hewes JL, Sharoff R, Song K, Norton EB, Kolls JK, McCombs JE. Vaccine-elicited IL-1R signaling results in Th17 TRM-mediated immunity. Commun Biol 2024; 7:433. [PMID: 38594380 PMCID: PMC11003962 DOI: 10.1038/s42003-024-06138-0] [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: 05/18/2023] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Lung tissue resident memory (TRM) cells are thought to play crucial roles in lung host defense. We have recently shown that immunization with the adjuvant LTA1 (derived from the A1 domain of E. coli heat labile toxin) admixed with OmpX from K. pneumoniae can elicit antigen specific lung Th17 TRM cells that provide serotype independent immunity to members of the Enterobacteriaceae family. However, the upstream requirements to generate these cells are unclear. Single-cell RNA-seq showed that vaccine-elicited Th17 TRM cells expressed high levels of IL-1R1, suggesting that IL-1 family members may be critical to generate these cells. Using a combination of genetic and antibody neutralization approaches, we show that Th17 TRM cells can be generated independent of caspase-1 but are compromised when IL-1α is neutralized. Moreover IL-1α could serve as a molecular adjuvant to generate lung Th17 TRM cells independent of LTA1. Taken together, these data suggest that IL-1α plays a major role in vaccine-mediated lung Th17 TRM generation.
Collapse
Affiliation(s)
- Joseph P Hoffmann
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Akhilesh Srivastava
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Haoran Yang
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Naoki Iwanaga
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki, Japan
| | - T Parks Remcho
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jenny L Hewes
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Rayshma Sharoff
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kejing Song
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Elizabeth B Norton
- Department of Immunology and Microbiology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jay K Kolls
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA
| | - Janet E McCombs
- Center for Translational Research in Infection and Inflammation, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
3
|
Yang YS, Chen HJ, Chen XC, Tang HJ, Chang FJ, Huang YL, Pan YL, Kesavan DK, Chen HY, Shang HS, Kuo SC, Chen TL, Chiang MH. Elizabethkingia anophelis outer membrane vesicles as a novel vaccine candidate against infection: insights into immune response and potential for passive immunity. mSphere 2023; 8:e0040023. [PMID: 38014949 PMCID: PMC10732079 DOI: 10.1128/msphere.00400-23] [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: 07/17/2023] [Accepted: 10/13/2023] [Indexed: 11/29/2023] Open
Abstract
IMPORTANCE Elizabethkingia anophelis, a Gram-negative pathogen, causes infections such as bacteraemia, pneumonia, and neonatal meningitis. The pathogen resists most antimicrobial classes, making novel approaches urgently needed. In natural settings, Gram-negative bacteria secrete outer membrane vesicles (OMVs) that carry important molecules in the bacterial life cycle. These OMVs are enriched with proteins involved in virulence, survival, and carbohydrate metabolism, making them a promising source for vaccine development against the pathogen. This study investigated the efficacy of imipenem-induced OMVs (iOMVs) as a vaccine candidate against E. anophelis infection in a mouse pneumonia model. Mice immunized with iOMVs were completely protected during lethal-dose challenges. Passive immunization with hyperimmune sera and splenocytes conferred protection against lethal pneumonia. Further investigation is needed to understand the mechanisms underlying the protective effects of iOMV-induced passive immunity, such as the action on specific antibody subclasses or T cell subsets.
Collapse
Grants
- 109-2320-B-016-002-MY2, 110-2320-B-016-014, 111-2320-B-016-015, 112-2314-B-016-023, 112-2314-B-016-039, 112-2314-B-016-024-MY2 Ministry of Science and Technology, Taiwan (MOST)
- TSGH-E-111244, TSGH-E-112253 Tri-Service General Hospital (TSGH)
- CMNDMC11108, CMNDMC11206 Chi Mei Medical Center
- MND-MAB-110-049, MND-MAB-D-111072, MND-MAB-D-112115, MND-MAB-D-113078 MOD | Medical Affairs Bureau (MAB)
Collapse
Affiliation(s)
- Ya-Sung Yang
- Department of Internal Medicine, Division of Infectious Diseases and Tropical Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Jui Chen
- Department of Infectious Diseases, Chi Mei Medical Center, Tainan, Taiwan
| | - Xiao-Chun Chen
- Department and Graduate institute of Microbiology and Immunology, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Jen Tang
- Department of Infectious Diseases, Chi Mei Medical Center, Tainan, Taiwan
| | - Fang-Ju Chang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yun-Ling Huang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Ling Pan
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Dinesh Kumar Kesavan
- School of Material Science, Nanyang Technological University, Singapore, Singapore
| | - Huan-Yuan Chen
- Inflammation Core Facility, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Hung-Sheng Shang
- Department of Pathology, Division of Clinical Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shu-Chen Kuo
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Taiwan
| | - Te-Li Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Hsien Chiang
- Department and Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
4
|
Nielsen TB, Yan J, Slarve M, Li R, Junge JA, Luna BM, Wilkinson I, Yerramalla U, Spellberg B. Development of a Bispecific Antibody Targeting Clinical Isolates of Acinetobacter baumannii. J Infect Dis 2023; 227:1042-1049. [PMID: 36617220 PMCID: PMC10319980 DOI: 10.1093/infdis/jiac499] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/16/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND We previously reported developing 2 anticapsular monoclonal antibodies (mAbs) as a novel therapy for Acinetobacter baumannii infections. We sought to determine whether a bispecific mAb (bsAb) could improve avidity and efficacy while maximizing strain coverage in one molecule. METHODS Humanized mAb 65 was cloned into a single-chain variable fragment and attached to humanized mAb C8, combining their paratopes into a single bsAb (C73). We tested bsAb C73's strain coverage, binding affinity, ex vivo opsonic activity, and in vivo efficacy compared to each mAb alone and combined. RESULTS The bsAb demonstrated strain coverage, binding affinity, opsonization, and in vivo efficacy superior to either original mAb alone or combined. CONCLUSIONS A humanized bsAb targeting distinct A. baumannii capsule moieties enabled potent and effective coverage of disparate A. baumannii clinical isolates. The bsAb enhances feasibility of development by minimizing the number of components of a promising novel therapeutic for these difficult-to-treat infections.
Collapse
Affiliation(s)
- Travis B Nielsen
- Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
- Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, Illinois, USA
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Jun Yan
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Matthew Slarve
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Rachel Li
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Jason A Junge
- Translational Imaging Center, School of Engineering, University of Southern California, Los Angeles, California, USA
| | - Brian M Luna
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | | | | | - Brad Spellberg
- Los Angeles County + University of Southern California Medical Center, Los Angeles, California, USA
| |
Collapse
|
5
|
Zhu Y, Han Y, Almuntashiri S, Dutta S, Wang X, Owen CA, Zhang D. Dysregulation of miR-103a Mediates Cigarette Smoking-induced Lipid-laden Macrophage Formation. Am J Respir Cell Mol Biol 2022; 67:695-707. [PMID: 36066909 PMCID: PMC9743184 DOI: 10.1165/rcmb.2022-0202oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/02/2022] [Indexed: 12/15/2022] Open
Abstract
Cigarette smoke (CS) is considered a major risk factor for chronic obstructive pulmonary disease (COPD) that is currently the third leading cause of death in the United States. Studies have indicated that patients with COPD have elevated blood low-density lipoprotein levels, which may contribute to the dysregulation of lipid metabolism. Accumulating data show that microRNAs (miRNAs) are involved in various human diseases. However, the role of microRNAs in the pathogenesis of COPD remains poorly defined. In this study, we found that miR-103a expression was significantly reduced in alveolar macrophages from smokers and patients with COPD versus that in alveolar macrophages from nonsmokers. Our data indicated that reactive oxygen species negatively regulate miR-103a in macrophages. Functionally, miR-103a modulates the expressions of genes involved in lipid metabolism and directly targets low-density lipoprotein receptors in macrophages. Furthermore, overexpression of miR-103a suppressed the accumulation of lipid droplets and reduced the reactive oxygen species, both in vitro and in vivo. Taken together, our findings indicate that downregulation of miR-103a contributes to cigarette smoke-induced lipid-laden macrophage formation and plays a critical role in lipid homeostasis in lung macrophages in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Yohan Han
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Sultan Almuntashiri
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
- Department of Clinical Pharmacy, College of Pharmacy, University of Hail, Hail, Saudi Arabia
| | - Saugata Dutta
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Xiaoyun Wang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Caroline A. Owen
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
6
|
Huo W, Busch LM, Hernandez-Bird J, Hamami E, Marshall CW, Geisinger E, Cooper VS, van Opijnen T, Rosch JW, Isberg RR. Immunosuppression broadens evolutionary pathways to drug resistance and treatment failure during Acinetobacter baumannii pneumonia in mice. Nat Microbiol 2022; 7:796-809. [PMID: 35618774 PMCID: PMC9159950 DOI: 10.1038/s41564-022-01126-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 04/20/2022] [Indexed: 01/02/2023]
Abstract
Acinetobacter baumannii is increasingly refractory to antibiotic treatment in healthcare settings. As is true of most human pathogens, the genetic path to antimicrobial resistance (AMR) and the role that the immune system plays in modulating AMR during disease are poorly understood. Here we reproduced several routes to fluoroquinolone resistance, performing evolution experiments using sequential lung infections in mice that are replete with or depleted of neutrophils, providing two key insights into the evolution of drug resistance. First, neutropenic hosts acted as reservoirs for the accumulation of drug resistance during drug treatment. Selection for variants with altered drug sensitivity profiles arose readily in the absence of neutrophils, while immunocompetent animals restricted the appearance of these variants. Secondly, antibiotic treatment failure in the immunocompromised host was shown to occur without clinically defined resistance, an unexpected result that provides a model for how antibiotic failure occurs clinically in the absence of AMR. The genetic mechanism underlying both these results is initiated by mutations activating the drug egress pump regulator AdeL, which drives persistence in the presence of antibiotic. Therefore, antibiotic persistence mutations present a two-pronged risk during disease, causing drug treatment failure in the immunocompromised host while simultaneously increasing the emergence of high-level AMR.
Collapse
Affiliation(s)
- Wenwen Huo
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Lindsay M Busch
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | - Juan Hernandez-Bird
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Efrat Hamami
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA
| | - Christopher W Marshall
- Department of Microbiology and Molecular Genetics and Center for Evolutionary Biology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Biological Sciences, Marquette University, Milwaukee, WI, USA
| | | | - Vaughn S Cooper
- Department of Microbiology and Molecular Genetics and Center for Evolutionary Biology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Jason W Rosch
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Ralph R Isberg
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
7
|
Kim S, Fesenmeier DJ, Park S, Torregrosa-Allen SE, Elzey BD, Won YY. Pulmonary Pharmacokinetics of Polymer Lung Surfactants Following Pharyngeal Administration in Mice. Biomacromolecules 2022; 23:2471-2484. [PMID: 35580262 DOI: 10.1021/acs.biomac.2c00221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We have recently discovered that pulmonary administration of nanoparticles (micelles) formed by amphiphilic poly(styrene-block-ethylene glycol) (PS-PEG) block copolymers has the potential to treat a lung disorder involving lung surfactant (LS) dysfunction (called acute respiratory distress syndrome (ARDS)), as PS-PEG nanoparticles are capable of reducing the surface tension of alveolar fluid, while they are resistant to deactivation caused by plasma proteins/inflammation products unlike natural LS. Herein, we report studies of the clearance pathways and kinetics of PS-PEG nanoparticles from the lung, which are essential for designing further preclinical IND-enabling studies. Using fluorescently labeled PS-PEG nanoparticles, we found that, following pharyngeal aspiration in mice, the retention of these nanoparticles in the lungs extends over 2 weeks, while their transport into other (secondary) organs is relatively insignificant. An analysis based on a multicompartmental pharmacokinetic model suggests a biphasic mechanism involving a fast mucociliary escalator process through the conducting airways and much slower alveolar clearance processes by the action of macrophages and also via direct translocation into the circulation. An excessive dose of PS-PEG nanoparticles led to prolonged retention in the lungs due to saturation of the alveolar clearance capacity.
Collapse
Affiliation(s)
- Seyoung Kim
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Daniel J Fesenmeier
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sungwan Park
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sandra E Torregrosa-Allen
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| | - Bennett D Elzey
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| | - You-Yeon Won
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, Indiana 47907, United States.,Purdue University Center for Cancer Research, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
8
|
Robinson EK, Worthington A, Poscablo D, Shapleigh B, Salih MM, Halasz H, Seninge L, Mosqueira B, Smaliy V, Forsberg EC, Carpenter S. lincRNA-Cox2 Functions to Regulate Inflammation in Alveolar Macrophages during Acute Lung Injury. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1886-1900. [PMID: 35365562 PMCID: PMC9038212 DOI: 10.4049/jimmunol.2100743] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 02/05/2022] [Indexed: 12/15/2022]
Abstract
Our respiratory system is vital to protect us from the surrounding nonsterile environment; therefore, it is critical for a state of homeostasis to be maintained through a balance of inflammatory cues. Recent studies have shown that actively transcribed noncoding regions of the genome are emerging as key regulators of biological processes, including inflammation. lincRNA-Cox2 is one such example of an inflammatory inducible long intergenic noncoding RNA functioning to fine-tune immune gene expression. Using bulk and single-cell RNA sequencing, in addition to FACS, we find that lincRNA-Cox2 is most highly expressed in the lung and is most upregulated after LPS-induced lung injury (acute lung injury [ALI]) within alveolar macrophages, where it functions to regulate inflammation. We previously reported that lincRNA-Cox2 functions to regulate its neighboring protein Ptgs2 in cis, and in this study, we use genetic mouse models to confirm its role in regulating gene expression more broadly in trans during ALI. Il6, Ccl3, and Ccl5 are dysregulated in the lincRNA-Cox2-deficient mice and can be rescued to wild type levels by crossing the deficient mice with our newly generated lincRNA-Cox2 transgenic mice, confirming that this gene functions in trans. Many genes are specifically regulated by lincRNA-Cox2 within alveolar macrophages originating from the bone marrow because the phenotype can be reversed by transplantation of wild type bone marrow into the lincRNA-Cox2-deficient mice. In conclusion, we show that lincRNA-Cox2 is a trans-acting long noncoding RNA that functions to regulate immune responses and maintain homeostasis within the lung at baseline and on LPS-induced ALI.
Collapse
Affiliation(s)
- Elektra Kantzari Robinson
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA
| | - Atesh Worthington
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; and
| | - Donna Poscablo
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; and
| | - Barbara Shapleigh
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA
| | - Mays Mohammed Salih
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA
| | - Haley Halasz
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA
| | - Lucas Seninge
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA
| | - Benny Mosqueira
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA
| | - Valeriya Smaliy
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA
| | - E Camilla Forsberg
- Institute for the Biology of Stem Cells, University of California-Santa Cruz, Santa Cruz, CA; and
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA
| | - Susan Carpenter
- Department of Molecular, Cell and Developmental Biology, University of California Santa Cruz, Santa Cruz, CA;
| |
Collapse
|
9
|
Murine Respiratory Tract Infection with Classical Klebsiella pneumoniae Induces Bronchus-Associated Lymphoid Tissue. Infect Immun 2022; 90:e0059621. [PMID: 35311545 DOI: 10.1128/iai.00596-21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Klebsiella pneumoniae is a Gram-negative, opportunistic pathogen that commonly causes nosocomial pneumonia, urinary tract infection, and septicemia. Our recent work utilizing a murine model of respiratory tract infection with classical K. pneumoniae demonstrated leukocyte aggregates in the lungs of mice at 28 days postinfection. Here, we sought to characterize the composition and development of these structures. Histopathological analyses of murine lungs revealed immune cell clusters surrounding the pulmonary vasculature and airways by 14 days postinfection, resembling inducible bronchus-associated lymphoid tissue (iBALT). Further investigation of these structures demonstrated central B cell aggregates with concomitant dispersed T cells. At day 28 postinfection, these lymphoid clusters expressed germinal center markers and CXCL12, qualifying these structures as iBALT with nonclassical B cell follicles. Investigations in mutant mice revealed that those lacking B and/or T cells were not able to form fully defined iBALT structures, although some rudimentary B cell clusters were identified in mice lacking T cells. The longevity of K. pneumoniae-induced BALT was assessed for up to 120 days postinfection. Lymphoid aggregates significantly decreased in size and quantity by 90 days after K. pneumoniae infection; however, aggregates persisted in mice that were restimulated with K. pneumoniae every 30 days. Finally, infections of mice with an array of classical K. pneumoniae clinical isolates demonstrated that the development of these structures is a common feature of K. pneumoniae lung infection. Together, these data confirm that murine lungs infected with K. pneumoniae develop iBALT, which may play a role in pulmonary immunity to this troublesome pathogen.
Collapse
|
10
|
Abstract
Background: Extremely drug-resistant (XDR) Acinetobacter baumannii is a notorious and frequently encountered pathogen demanding novel therapeutic interventions. An initial monoclonal antibody (MAb), C8, raised against A. baumannii capsule proved a highly effective treatment against a minority of clinical isolates. To overcome this limitation, we broadened coverage by developing a second antibody for use in a combination regimen. Methods: We sought to develop an additional anti-A. baumannii MAb through hybridoma technology by immunizing mice with sublethal inocula of virulent, XDR clinical isolates not bound by MAb C8. Results: We identified a new antibacterial MAb, 65, which bound to strains in a pattern distinct from and complementary to MAb C8. MAb 65 enhanced macrophage opsonophagocytosis of targeted strains and markedly improved survival in lethal bacteremic sepsis and aspiration pneumonia murine models of A. baumannii infection. MAb 65 was also synergistic with colistin, substantially enhancing protection compared to monotherapy. Treatment with MAb 65 significantly reduced blood bacterial density, ameliorated cytokine production (IL-1β, IL-6, IL-10, and TNF), and sepsis biomarkers. Conclusions: We describe a novel MAb targeting A. baumannii that broadens immunotherapeutic strain coverage, is highly potent and effective, and synergistically improves outcomes in combination with antibiotics.
Collapse
|
11
|
FDA Public Workshop Summary: Advancing Animal Models for Antibacterial Drug Development. Antimicrob Agents Chemother 2020; 65:AAC.01983-20. [PMID: 33106262 DOI: 10.1128/aac.01983-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The U.S. Food and Drug Administration (FDA) hosted a public workshop entitled "Advancing Animal Models for Antibacterial Drug Development" on 5 March 2020. The workshop mainly focused on models of pneumonia caused by Pseudomonas aeruginosa and Acinetobacter baumannii The program included discussions from academic investigators, industry, and U.S. government scientists. The potential use of mouse, rabbit, and pig models for antibacterial drug development was presented and discussed.
Collapse
|
12
|
Felgenhauer JL, Brune JE, Long ME, Manicone AM, Chang MY, Brabb TL, Altemeier WA, Frevert CW. Evaluation of Nutritional Gel Supplementation in C57BL/6J Mice Infected with Mouse-Adapted Influenza A/PR/8/34 Virus. Comp Med 2020; 70:471-486. [PMID: 33323164 PMCID: PMC7754200 DOI: 10.30802/aalas-cm-20-990138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 11/05/2019] [Accepted: 09/04/2020] [Indexed: 12/11/2022]
Abstract
Mice are a common animal model for the study of influenza virus A (IAV). IAV infection causes weight loss due to anorexia and dehydration, which can result in early removal of mice from a study when they reach a humane endpoint. To reduce the number of mice prematurely removed from an experiment, we assessed nutritional gel (NG) supplementation as a support strategy for mice infected with mouse-adapted Influenza A/Puerto Rico/8/34 (A/PR/8/34; H1N1) virus. We hypothesized that, compared with the standard of care (SOC), supplementation with NG would reduce weight loss and increase survival in mice infected with IAV without impacting the initial immune response to infection. To assess the effects of NG, male and female C57BL/6J mice were infected with IAV at low, intermediate, or high doses. When compared with SOC, mice given NG showed a significant decrease in the maximal percent weight loss at all viral doses in males and at the intermediate dose for females. Mice supplemented with NG had no deaths for either sex at the intermediate dose and a significant increase in survival in males at the high viral dose. Supplementation with NG did not alter the viral titer or the pulmonary recruitment of immune cells as measured by cell counts and flow cytometry of cells recovered in bronchoalveolar lavage (BAL) fluid in either sex. However, mice given NG had a significant reduction in IL6 and TNFα in BAL fluid and no significant differences in CCL2, IL4, IL10, CXCL1, CXCL2, and VEGF. The results of this study show that as compared with infected SOC mice, infected mice supplemented with NG have reduced weight loss and increased survival, with males showing a greater benefit. These results suggest that NG should be considered as a support strategy and indicate that sex is an important biologic variable in mice infected with IAV.
Collapse
Key Words
- iav, influenza a virus
- soc, standard of care
- ng, nutritional gel
- eud50, euthanasia dose 50
- ld50, lethal-dose 50
- pfu, plaque forming unit
- dpi, days post infection
- il6, interleukin 6
- sem, standard error of mean
- ns, no significance
Collapse
Affiliation(s)
- Jessica L Felgenhauer
- Department of Comparative Medicine, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington
| | - Jourdan E Brune
- Department of Comparative Medicine, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington
| | - Matthew E Long
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington
| | - Anne M Manicone
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington
| | - Mary Y Chang
- Department of Comparative Medicine, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington
| | - Thea L Brabb
- Department of Comparative Medicine, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington
| | - William A Altemeier
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington
| | - Charles W Frevert
- Department of Comparative Medicine, University of Washington, Seattle, Washington; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, Washington; Center for Lung Biology, University of Washington at South Lake Union, Seattle, Washington;,
| |
Collapse
|
13
|
Hu X, Cai Y, Wang Y, Wang R, Wang J, Zhang B. Imaging of bioluminescent Klebsiella pneumoniae induced pulmonary infection in an immunosuppressed mouse model. J Int Med Res 2020; 48:300060520956473. [PMID: 33044099 PMCID: PMC7556177 DOI: 10.1177/0300060520956473] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Objective To establish a mouse model of bioluminescent Klebsiella pneumoniae-induced lung infection, under different infection states after pretreatment with various dosages of cyclophosphamide (CTX). Methods A K. pneumoniae strain carrying the luxCDABE operon was used to infect immunocompetent mice (intraperitoneal injection of saline at 4 days and 1 day prior to experimental lung infection) and immunodeficient mice (50 mg/kg CTX at 4 days and 50 mg/kg CTX at 1 day prior to lung infection; or 150 mg/kg CTX at 4 days and 100 mg/kg CTX at 1 day prior to lung infection). Disease progression was monitored in living mice using a bioluminescence imaging system. The bioluminescent images, bacterial loads in lungs, blood cytological changes and histopathology of lungs were analysed. Results K. pneumoniae-induced lung infection models were established in mice pretreated with CTX. Different doses of CTX led to different severities of lung infection. Mice pretreated with 150/100 mg/kg CTX were more suitable for real-time monitoring as they had more typical bioluminescent images of lung infection, more obvious changes in the bioluminescent intensity values, more bacterial colonies in the lungs and more distinct pulmonary pathological changes. Conclusions A stable bioluminescent K. pneumonia-induced lung infection model was successfully established in mice pretreated with CTX, which can be semi-quantitatively monitored in real-time.
Collapse
Affiliation(s)
- Xing Hu
- Medical School of Chinese PLA, Beijing, China
| | - Yun Cai
- Medical School of Chinese PLA, Beijing, China.,Department of Pharmacy, Centre of Medicine Clinical Research, PLA General Hospital, Beijing, China
| | - Yuhang Wang
- Medical School of Chinese PLA, Beijing, China
| | - Rui Wang
- Department of Pharmacy, Centre of Medicine Clinical Research, PLA General Hospital, Beijing, China
| | - Jin Wang
- Department of Pharmacy, Centre of Medicine Clinical Research, PLA General Hospital, Beijing, China
| | - Bo Zhang
- Medical School of Chinese PLA, Beijing, China.,Department of Respiratory Medicine, Air Force General Hospital, PLA, Beijing, China
| |
Collapse
|
14
|
New Approach Methods to Evaluate Health Risks of Air Pollutants: Critical Design Considerations for In Vitro Exposure Testing. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17062124. [PMID: 32210027 PMCID: PMC7143849 DOI: 10.3390/ijerph17062124] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/11/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022]
Abstract
Air pollution consists of highly variable and complex mixtures recognized as major contributors to morbidity and mortality worldwide. The vast number of chemicals, coupled with limitations surrounding epidemiological and animal studies, has necessitated the development of new approach methods (NAMs) to evaluate air pollution toxicity. These alternative approaches include in vitro (cell-based) models, wherein toxicity of test atmospheres can be evaluated with increased efficiency compared to in vivo studies. In vitro exposure systems have recently been developed with the goal of evaluating air pollutant-induced toxicity; though the specific design parameters implemented in these NAMs-based studies remain in flux. This review aims to outline important design parameters to consider when using in vitro methods to evaluate air pollutant toxicity, with the goal of providing increased accuracy, reproducibility, and effectiveness when incorporating in vitro data into human health evaluations. This review is unique in that experimental considerations and lessons learned are provided, as gathered from first-hand experience developing and testing in vitro models coupled to exposure systems. Reviewed design aspects include cell models, cell exposure conditions, exposure chambers, and toxicity endpoints. Strategies are also discussed to incorporate in vitro findings into the context of in vivo toxicity and overall risk assessment.
Collapse
|
15
|
Luna BM, Yan J, Reyna Z, Moon E, Nielsen TB, Reza H, Lu P, Bonomo R, Louie A, Drusano G, Bulitta J, She R, Spellberg B. Natural history of Acinetobacter baumannii infection in mice. PLoS One 2019; 14:e0219824. [PMID: 31318907 PMCID: PMC6638954 DOI: 10.1371/journal.pone.0219824] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/03/2019] [Indexed: 01/30/2023] Open
Abstract
In 2017, the WHO identified Acinetobacter baumannii as the top priority for the development of new antibiotics. Despite the need for new antibiotics, there remains a lack of well validated preclinical tools for A. baumannii. Here, we characterize and validate a mouse model for A. baumannii translational research. Antibiotic sensitivity for meropenem, amikacin, and polymyxin b was determined by the broth microdilution MIC assay. LD100 inoculums, in both blood and lung infection models, were determined in male and female C3HeB/FeJ mice that were challenged with various A. baumannii clinical isolates. Blood (blood infection model) or blood and lung tissue (lung infection model) were collected from infected mice at 2 and 18 hours and the bacterial burden was determined by quantitative culture. Blood chemistry was analyzed using the iStat system. Cytokines (IL-1ß, TNF, IL-6, and IL-10) were measured in the blood and lung homogenate by ELISA assay. Lung sections (H&E stains) were scored by a pathologist. In the blood infection model, the cytokines and physiological data indicate that mice become moribund due to sepsis (low blood pH, falling bicarbonate, and a rising base deficit), whereas mice become moribund due to respiratory failure (low blood pH, rising bicarbonate, and a falling base deficit) in the oral aspiration pneumonia model. We also characterized the timing of changes in various clinical and biomarker endpoints, which can serve as a basis for future interventional studies. Susceptibility was generally similar across gender and infection route. However, we did observe that female mice were approximately 2-fold more sensitive to LAC-4 ColR in the blood infection model. We also observed that female mice were more than 10-fold more resistant to VA-AB41 in the oral aspiration pneumonia model. These results establish parameters to follow in order to assess efficacy of novel preventative and therapeutic approaches for these infections.
Collapse
Affiliation(s)
- Brian M. Luna
- Department of Medicine, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
- Department of Molecular Microbiology and Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
- * E-mail:
| | - Jun Yan
- Department of Medicine, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
- Department of Molecular Microbiology and Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
| | - Zeferino Reyna
- Department of Pathology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
| | - Eugene Moon
- Department of Medicine, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
- Department of Molecular Microbiology and Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
| | - Travis B. Nielsen
- Department of Medicine, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
- Department of Molecular Microbiology and Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
| | - Hernan Reza
- Department of Medicine, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
- Department of Molecular Microbiology and Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
| | - Peggy Lu
- Department of Medicine, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
- Department of Molecular Microbiology and Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
| | - Robert Bonomo
- Departments of Medicine, Pharmacology, and Molecular Biology and Microbiology, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Arnold Louie
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, United States of America
| | - George Drusano
- Institute for Therapeutic Innovation, College of Medicine, University of Florida, Orlando, Florida, United States of America
| | - Jürgen Bulitta
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, United States of America
| | - Rosemary She
- Department of Pathology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
| | - Brad Spellberg
- Department of Medicine, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
- Department of Molecular Microbiology and Immunology, Keck School of Medicine at the University of Southern California (USC), Los Angeles, California, United States of America
| |
Collapse
|
16
|
May HC, Yu JJ, Zhang H, Wang Y, Cap AP, Chambers JP, Guentzel MN, Arulanandam BP. Thioredoxin-A is a virulence factor and mediator of the type IV pilus system in Acinetobacter baumannii. PLoS One 2019; 14:e0218505. [PMID: 31265467 PMCID: PMC6605650 DOI: 10.1371/journal.pone.0218505] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 06/04/2019] [Indexed: 01/08/2023] Open
Abstract
The Gram-negative pathogen, Acinetobacter baumannii has emerged as a global nosocomial health threat affecting the majority of hospitals in the U.S. and abroad. The redox protein thioredoxin has been shown to play several roles in modulation of cellular functions affecting various virulence factors in Gram-negative pathogens. This study aims to explore the role of thioredoxin-A protein (TrxA) in A. baumannii virulence. We determined that deletion of the TrxA gene did not significantly affect resistance to environmental stressors such as temperature, salt, and pH. However, TrxA was critical for survival in the presence of elevated levels of hydrogen peroxide. Lack of TrxA was associated with decreased expression of type IV pili related genes and an inability to undergo normal twitching motility. Interestingly, the TrxA-null mutant was able to form biofilms better than the wildtype (WT) and was observed to be significantly less virulent than the WT in a pulmonary infection model. These results are supportive of thioredoxin playing a key role in A. baumannii virulence.
Collapse
Affiliation(s)
- Holly C. May
- South Texas Center for Emerging Infectious Disease and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Jieh-Juen Yu
- South Texas Center for Emerging Infectious Disease and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Hao Zhang
- South Texas Center for Emerging Infectious Disease and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Yufeng Wang
- South Texas Center for Emerging Infectious Disease and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Andrew P. Cap
- Coagulation and Blood Research Program, US Army Institute for Surgical Research, JBSA Fort Sam Houston, San Antonio, Texas, United States of America
| | - James P. Chambers
- South Texas Center for Emerging Infectious Disease and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - M. Neal Guentzel
- South Texas Center for Emerging Infectious Disease and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
| | - Bernard P. Arulanandam
- South Texas Center for Emerging Infectious Disease and Department of Biology, University of Texas at San Antonio, San Antonio, Texas, United States of America
- * E-mail:
| |
Collapse
|