1
|
Wang L, Lu Y, Liu J, Wang S, Fei Z, Zhang K, Zhang D, Jin X. Gegen Qinlian tablets delay Alzheimer's disease progression via inhibiting glial neuroinflammation and remodeling gut microbiota homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155394. [PMID: 38569294 DOI: 10.1016/j.phymed.2024.155394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/11/2024] [Accepted: 01/24/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Current therapeutic agents for AD have limited efficacy and often induce undesirable side effects. Gegen Qinlian tablets (GGQLT) are a well-known clearingheat formula used in clinical treatment of inflammatory diseases. Based on traditional Chinese medicine (TCM) theory, the strategy of clearing-heat is then compatible with the treatment of AD. However, it remains unknown whether GGQLT can exert neuroprotective effects and alleviate neuroinflammation in AD. PURPOSE This study aimed to evaluate the anti-AD effects of GGQLT and to decipher its intricate mechanism using integrative analyses of network pharmacology, transcriptomic RNA sequencing, and gut microbiota. METHODS The ingredients of GGQLT were analyzed using HPLC-ESI-Q/TOF-MS. The AD model was established by bilateral injection of Aβ1-42 into the intracerebroventricular space of rats. The Morris water maze was used to evaluate the cognitive function of the AD rats. The long-term toxicity of GGQLT in rats was assessed by monitoring their body weights and pathological alterations in the liver and kidney. Reactive astrocytes and microglia were assessed by immunohistochemistry by labeling GFAP and Iba-1. The levels of inflammatory cytokines in the hippocampus were evaluated using ELISA kits, RT-PCR, and Western blot, respectively. The potential anti-AD mechanism was predicted by analyses of RNA-sequencing and network pharmacology. Western blot and immunohistochemistry were utilized to detect the phosphorylation levels of IκBα, NF-κB p65, p38, ERK and JNK. The richness and composition of gut bacterial and fungal microflora were investigated via 16S rRNA and ITS sequencing. RESULTS Typical ingredients of GGQLT were identified using HPLC-ESI-Q/TOF-MS. GGQLT significantly improved the cognitive function of AD rats by suppressing the activation of microglia and astrocytes, improving glial morphology, and reducing the neuroinflammatory reactions in the hippocampus. RNA-sequencing, network and experimental pharmacological studies demonstrated that GGQLT inhibited the activation of NF-κB/MAPK signaling pathways in the hippocampus. GGQLT could also restore abnormal gut bacterial and fungal homeostasis and no longer-term toxicity of GGQLT was observed. CONCLUSIONS Our findings, for the first time, demonstrate GGQLT exhibit anti-AD effects and is worthy of further exploration and development.
Collapse
Affiliation(s)
- Lin Wang
- School of Pharmacy, China Medical University, No.77 of Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Ye Lu
- School of Pharmacy, China Medical University, No.77 of Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Jiamei Liu
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Siyi Wang
- The 1st Clinical Department, China Medical University, Shenyang 110122, China
| | - Zepeng Fei
- School of Pharmacy, China Medical University, No.77 of Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Kaiwen Zhang
- School of Pharmacy, China Medical University, No.77 of Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Dongfang Zhang
- School of Pharmacy, China Medical University, No.77 of Puhe Road, Shenyang North New Area, Shenyang 110122, China.
| | - Xin Jin
- School of Pharmacy, China Medical University, No.77 of Puhe Road, Shenyang North New Area, Shenyang 110122, China.
| |
Collapse
|
2
|
Herbel SM, Moyon L, Christ M, Elsayed EM, Caffrey BE, Malmsheimer S, Grin I, Hoffmann K, Surmann K, Blankenburg S, Jung AL, Herkt CE, Borsò M, Bozdag B, Imhof A, Becker A, Wagner S, Bange G, Völker U, Bertrams W, Marsico A, Schmeck B. Screening for eukaryotic motifs in Legionella pneumophila reveals Smh1 as bacterial deacetylase of host histones. Virulence 2022; 13:2042-2058. [PMID: 36411449 PMCID: PMC9704406 DOI: 10.1080/21505594.2022.2149973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Legionella pneumophila (L.p.) is a bacterial pathogen which is a common causative agent of pneumonia. In humans, it infects alveolar macrophages and transfers hundreds of virulence factors that interfere with cellular signalling pathways and the transcriptomic landscape to sustain its own replication. By this interaction, it has acquired eukaryote-like protein motifs by gene transfer events that partake in the pathogenicity of Legionella. In a computational screening approach for eukaryotic motifs in the transcriptome of Legionella, we identified the L.p. strain Corby protein ABQ55614 as putative histone-deacetylase and named it "suppressing modifier of histones 1" (Smh1). During infection, Smh1 is translocated from the Legionella vacuole into the host cytosol. When expressed in human macrophage THP-1 cells, Smh1 was localized predominantly in the nucleus, leading to broad histone H3 and H4 deacetylation, blunted expression of a large number of genes (e.g. IL-1β and IL-8), and fostered intracellular bacterial replication. L.p. with a Smh1 knockdown grew normally in media but showed a slight growth defect inside the host cell. Furthermore, Smh1 showed a very potent histone deacetylation activity in vitro, e.g. at H3K14, that could be inhibited by targeted mutation of the putative catalytic center inferred by analogy with eukaryotic HDAC8, and with the deacetylase inhibitor trichostatin A. In summary, Smh1 displays functional homology with class I/II type HDACs. We identified Smh1 as a new Legionella virulence factor with a eukaryote-like histone-deacetylase activity that moderates host gene expression and might pave the way for further histone modifications.IMPORTANCELegionella pneumophila (L.p.) is a prominent bacterial pathogen, which is a common causative agent of pneumonia. In order to survive inside the host cell, the human macrophage, it profoundly interacts with host cell processes to advance its own replication. In this study, we identify a bacterial factor, Smh1, with yet unknown function as a host histone deacetylase. The activity of this factor in the host cell leads to attenuated gene expression and increased intracellular bacterial replication.
Collapse
Affiliation(s)
- Stefanie M. Herbel
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Lambert Moyon
- Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Marvin Christ
- Department of Chemistry, Philipps-University Marburg, Marburg, Germany,Center for Synthetic Microbiology (SYNMIKRO), Philipps-University Marburg, Marburg, Germany
| | - Eslam M. Elsayed
- Center for Synthetic Microbiology (SYNMIKRO), Philipps-University Marburg, Marburg, Germany,Department of Biology, Philipps-Universität Marburg, Marburg, Germany,Department of Microbiology and Immunology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Brian E. Caffrey
- Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Silke Malmsheimer
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Tübingen, Germany
| | - Iwan Grin
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Tübingen, Germany
| | - Kerstin Hoffmann
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Kristin Surmann
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Sascha Blankenburg
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Anna Lena Jung
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Christina E. Herkt
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Marco Borsò
- Zentrallabor für Proteinanalytik, BioMedical Center, Faculty of Medicine, Ludwig-Maximilians-University of Munich, Planegg-Martinsried
| | - Beyza Bozdag
- Zentrallabor für Proteinanalytik, BioMedical Center, Faculty of Medicine, Ludwig-Maximilians-University of Munich, Planegg-Martinsried
| | - Axel Imhof
- Zentrallabor für Proteinanalytik, BioMedical Center, Faculty of Medicine, Ludwig-Maximilians-University of Munich, Planegg-Martinsried
| | - Anke Becker
- Center for Synthetic Microbiology (SYNMIKRO), Philipps-University Marburg, Marburg, Germany,Department of Biology, Philipps-Universität Marburg, Marburg, Germany
| | - Samuel Wagner
- Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Tübingen, Germany,German Center for Infection Research (DZIF), Partner-site Tübingen, Tübingen, Germany
| | - Gert Bange
- Department of Chemistry, Philipps-University Marburg, Marburg, Germany,Center for Synthetic Microbiology (SYNMIKRO), Philipps-University Marburg, Marburg, Germany,Max-Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Wilhelm Bertrams
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany
| | - Annalisa Marsico
- Computational Health Center, Helmholtz Zentrum München, Neuherberg, Germany
| | - Bernd Schmeck
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center (UGMLC), German Center for Lung Research (DZL), Philipps-University Marburg, Marburg, Germany,Center for Synthetic Microbiology (SYNMIKRO), Philipps-University Marburg, Marburg, Germany,Department of Medicine, Pulmonary and Critical Care Medicine, University Medical Center Giessen and Marburg, Philipps-University, Member of the German Center for Lung Research (DZL), Marburg, Germany,Institute for Lung Health (ILH), Justus-Liebig-University, Giessen, Germany,Member of the German Center for Infectious Disease Research (DZIF), Marburg, Germany,CONTACT Bernd Schmeck
| |
Collapse
|
3
|
Lin MS, Zhong HY, Yim RLH, Chen QY, Du HL, He HQ, Lin K, Zhao P, Gao R, Gao F, Zhang MY. Pan-cancer analysis of oncogenic TNFAIP2 identifying its prognostic value and immunological function in acute myeloid leukemia. BMC Cancer 2022; 22:1068. [PMID: 36243694 PMCID: PMC9571470 DOI: 10.1186/s12885-022-10155-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 10/04/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tumor necrosis factor alpha-induced protein 2 (TNFAIP2), a TNFα-inducible gene, appears to participate in inflammation, immune response, hematopoiesis, and carcinogenesis. However, the potential role of TNFAIP2 in the development of acute myeloid leukemia (AML) remains unknow yet. Therefore, we aimed to study the biological role of TNFAIP2 in leukemogenesis. METHODS TNFAIP2 mRNA level, prognostic value, co-expressed genes, differentially expressed genes, DNA methylation, and functional enrichment analysis in AML patients were explored via multiple public databases, including UALCAN, GTEx portal, Timer 2.0, LinkedOmics, SMART, MethSurv, Metascape, GSEA and String databases. Data from The Cancer Genome Atlas (TCGA), Gene Expression Omnibus (GEO) and Beat AML database were used to determine the associations between TNFAIP2 expression and various clinical or genetic parameters of AML patients. Moreover, the biological functions of TNFAIP2 in AML were investigated through in vitro experiments. RESULTS By large-scale data mining, our study indicated that TNFAIP2 was differentially expressed across different normal and tumor tissues. TNFAIP2 expression was significantly increased in AML, particularly in French-American-British (FAB) classification M4/M5 patients, compared with corresponding control tissues. Overexpression of TNFAIP2 was an independent poor prognostic factor of overall survival (OS) and was associated with unfavorable cytogenetic risk and gene mutations in AML patients. DNA hypermethylation of TNFAIP2 at gene body linked to upregulation of TNFAIP2 and inferior OS in AML. Functional enrichment analysis indicated immunomodulation function and inflammation response of TNFAIP2 in leukemogenesis. Finally, the suppression of TNFAIP resulted in inhibition of proliferation by altering cell-cycle progression and increase of cell death by promoting early and late apoptosis in THP-1 and U937AML cells. CONCLUSION Collectively, the oncogenic TNFAIP2 can function as a novel biomarker and prognostic factor in AML patients. The immunoregulation function of TNFAIP2 warrants further validation in AML.
Collapse
Affiliation(s)
- Mei-Si Lin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611730, China
| | - Hui-Yun Zhong
- Sichuan Vocational College of Health and Rehabilitation, Zigong, 643000, China
| | - Rita Lok-Hay Yim
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong, SAR, China
| | - Qi-Yan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611730, China
| | - Hong-Ling Du
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611730, China
| | - Hao-Qi He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611730, China
| | - Ke Lin
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, 611730, China
| | - Peng Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611730, China
| | - Ru Gao
- Department of Nursing, Chengdu Wenjiang People's Hospital, Chengdu, 611100, Sichuan, China.
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611730, China.
| | - Min-Yue Zhang
- Division of Hematology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200127, China.
| |
Collapse
|
4
|
Jin G, Liu Y, Xu W, Li Y, Zhang H, Qiu S, Gao C, Liu S. Tnfaip2 promotes atherogenesis by enhancing oxidative stress induced inflammation. Mol Immunol 2022; 151:41-51. [PMID: 36084515 DOI: 10.1016/j.molimm.2022.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/09/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022]
Abstract
The inflammation is considered to be the crucial determinants of lesion progression and plaque stability during atherogenesis. Tnfaip2 appears to be a regulator for carcinogenesis and infectious diseases. But its role in atherosclerosis is not clear. Here we first report that Tnfaip2 promotes the formation of atherosclerosis through enhancing the inflammation under oxidative stress condition. Although the endogenous expression of Tnfaip2 was upregulated under oxidative stress condition, the overexpressed Tnfaip2 could promote cells proliferation. This might result from the ability of promoting cells entering G2/M phase. Conversely, the cells proliferation and migration were significantly reduced in Tnfaip2 knockdown cells through inhibiting the activation of NF-κB/MAPK/Akt signaling pathways. However, the efferocytosis increased markedly due to the upregulation of "eat me" receptors, such as CD36, SR-A, and SR-B1, and the downregulation of "don't eat me" signal CD47. As a consequence, Tnfaip2 deficiency in bone marrow-derived cells inhibited atherosclerosis development in Ldlr-/- mice fed a high-fat diet accompanied by decreased inflammatory cytokines and shTnfaip2 could reduce the plaque lesions in ApoE-/- mice. These results indicate that Tnfaip2 might play an important role during atherogenesis.
Collapse
Affiliation(s)
- Guiyuan Jin
- Department of Immunology, Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Ji'nan, PR China; Medical Research Centre, Affiliated Hospital of Ji'ning Medical University, Ji'ning, Shandong Province, PR China
| | - Ying Liu
- Department of Immunology, Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Ji'nan, PR China
| | - Wenwen Xu
- Department of Immunology, Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Ji'nan, PR China
| | - Yan Li
- Department of Blood Transfusion, Qilu Hospital of Shandong University, Ji'nan, PR China
| | - Heng Zhang
- Department of Immunology, Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Ji'nan, PR China
| | - Shuoke Qiu
- Department of Immunology, Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Ji'nan, PR China
| | - Chengjiang Gao
- Department of Immunology, Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Ji'nan, PR China
| | - Suxia Liu
- Department of Immunology, Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Ji'nan, PR China.
| |
Collapse
|
5
|
Hiyoshi M, Takahashi N, Eltalkhawy YM, Noyori O, Lotfi S, Panaampon J, Okada S, Tanaka Y, Ueno T, Fujisawa JI, Sato Y, Suzuki T, Hasegawa H, Tokunaga M, Satou Y, Yasunaga JI, Matsuoka M, Utsunomiya A, Suzu S. M-Sec induced by HTLV-1 mediates an efficient viral transmission. PLoS Pathog 2021; 17:e1010126. [PMID: 34843591 PMCID: PMC8659635 DOI: 10.1371/journal.ppat.1010126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/09/2021] [Accepted: 11/17/2021] [Indexed: 11/19/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1) infects target cells primarily through cell-to-cell routes. Here, we provide evidence that cellular protein M-Sec plays a critical role in this process. When purified and briefly cultured, CD4+ T cells of HTLV-1 carriers, but not of HTLV-1- individuals, expressed M-Sec. The viral protein Tax was revealed to mediate M-Sec induction. Knockdown or pharmacological inhibition of M-Sec reduced viral infection in multiple co-culture conditions. Furthermore, M-Sec knockdown reduced the number of proviral copies in the tissues of a mouse model of HTLV-1 infection. Phenotypically, M-Sec knockdown or inhibition reduced not only plasma membrane protrusions and migratory activity of cells, but also large clusters of Gag, a viral structural protein required for the formation of viral particles. Taken together, these results suggest that M-Sec induced by Tax mediates an efficient cell-to-cell viral infection, which is likely due to enhanced membrane protrusions, cell migration, and the clustering of Gag. In the present study, we identified the cellular protein M-Sec as a host factor necessary for de novo infection of human T-cell leukemia virus type 1 (HTLV-1), the causative retrovirus of an aggressive blood cancer known as adult T-cell leukemia/lymphoma. The inhibition or knockdown of M-Sec in infected cells resulted in a reduced viral infection in several culture models and a mouse model. We recently demonstrated a similar role of M-Sec in macrophages infected with another human retrovirus HIV-1, but it has been generally thought that M-Sec is not related to HTLV-1 infection because of the lack of its expression in CD4+ T cells, the major target of HTLV-1. In this study, we revealed that CD4+ T cells of HTLV-1 asymptomatic carriers, but not those of HTLV-1- individuals, expressed M-Sec, and that the viral protein Tax mediated the induction of M-Sec. Thus, M-Sec is a new and useful tool for further understanding the process of HTLV-1 transmission.
Collapse
Affiliation(s)
- Masateru Hiyoshi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
- * E-mail: (MH); (SS)
| | - Naofumi Takahashi
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Youssef M. Eltalkhawy
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Osamu Noyori
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Sameh Lotfi
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Jutatip Panaampon
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Seiji Okada
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Yuetsu Tanaka
- School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Takaharu Ueno
- Department of Microbiology, Kansai Medical University, Osaka, Japan
| | | | - Yuko Sato
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hideki Hasegawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masahito Tokunaga
- Department of Hematology, Imamura General Hospital, Kagoshima, Japan
| | - Yorifumi Satou
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
| | - Jun-ichirou Yasunaga
- Department of Hematology, Rheumatology and Infectious Diseases, Kumamoto University School of Medicine, Kumamoto, Japan
| | - Masao Matsuoka
- Department of Hematology, Rheumatology and Infectious Diseases, Kumamoto University School of Medicine, Kumamoto, Japan
| | - Atae Utsunomiya
- Department of Hematology, Imamura General Hospital, Kagoshima, Japan
- Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Shinya Suzu
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- * E-mail: (MH); (SS)
| |
Collapse
|
6
|
Halwe S, Kupke A, Vanshylla K, Liberta F, Gruell H, Zehner M, Rohde C, Krähling V, Gellhorn Serra M, Kreer C, Klüver M, Sauerhering L, Schmidt J, Cai Z, Han F, Young D, Yang G, Widera M, Koch M, Werner A, Kämper L, Becker N, Marlow MS, Eickmann M, Ciesek S, Schiele F, Klein F, Becker S. Intranasal Administration of a Monoclonal Neutralizing Antibody Protects Mice against SARS-CoV-2 Infection. Viruses 2021; 13:v13081498. [PMID: 34452363 PMCID: PMC8402634 DOI: 10.3390/v13081498] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/24/2021] [Accepted: 07/25/2021] [Indexed: 12/18/2022] Open
Abstract
Despite the recent availability of vaccines against severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), there is an urgent need for specific anti-SARS-CoV-2 drugs. Monoclonal neutralizing antibodies are an important drug class in the global fight against the SARS-CoV-2 pandemic due to their ability to convey immediate protection and their potential to be used as both prophylactic and therapeutic drugs. Clinically used neutralizing antibodies against respiratory viruses are currently injected intravenously, which can lead to suboptimal pulmonary bioavailability and thus to a lower effectiveness. Here we describe DZIF-10c, a fully human monoclonal neutralizing antibody that binds the receptor-binding domain of the SARS-CoV-2 spike protein. DZIF-10c displays an exceptionally high neutralizing potency against SARS-CoV-2, retains full activity against the variant of concern (VOC) B.1.1.7 and still neutralizes the VOC B.1.351, although with reduced potency. Importantly, not only systemic but also intranasal application of DZIF-10c abolished the presence of infectious particles in the lungs of SARS-CoV-2 infected mice and mitigated lung pathology when administered prophylactically. Along with a favorable pharmacokinetic profile, these results highlight DZIF-10c as a novel human SARS-CoV-2 neutralizing antibody with high in vitro and in vivo antiviral potency. The successful intranasal application of DZIF-10c paves the way for clinical trials investigating topical delivery of anti-SARS-CoV-2 antibodies.
Collapse
Affiliation(s)
- Sandro Halwe
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Alexandra Kupke
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Kanika Vanshylla
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (K.V.); (H.G.); (M.Z.); (C.K.); (F.K.)
| | - Falk Liberta
- Biotherapeutics Discovery, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88397 Biberach an der Riss, Germany; (F.L.); (F.S.)
| | - Henning Gruell
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (K.V.); (H.G.); (M.Z.); (C.K.); (F.K.)
| | - Matthias Zehner
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (K.V.); (H.G.); (M.Z.); (C.K.); (F.K.)
| | - Cornelius Rohde
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Verena Krähling
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Michelle Gellhorn Serra
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Christoph Kreer
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (K.V.); (H.G.); (M.Z.); (C.K.); (F.K.)
| | - Michael Klüver
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Lucie Sauerhering
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Jörg Schmidt
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
| | - Zheng Cai
- Biotherapeutics Molecule Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877, USA; (Z.C.); (F.H.); (D.Y.); (G.Y.); (M.S.M.)
| | - Fei Han
- Biotherapeutics Molecule Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877, USA; (Z.C.); (F.H.); (D.Y.); (G.Y.); (M.S.M.)
| | - David Young
- Biotherapeutics Molecule Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877, USA; (Z.C.); (F.H.); (D.Y.); (G.Y.); (M.S.M.)
| | - Guangwei Yang
- Biotherapeutics Molecule Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877, USA; (Z.C.); (F.H.); (D.Y.); (G.Y.); (M.S.M.)
| | - Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60596 Frankfurt am Main, Germany; (M.W.); (S.C.)
| | - Manuel Koch
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany;
- Institute for Dental Research and Oral Musculoskeletal Biology and Center for Biochemistry, University of Cologne, 50931 Cologne, Germany
| | - Anke Werner
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
| | - Lennart Kämper
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
| | - Nico Becker
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
| | - Michael S. Marlow
- Biotherapeutics Molecule Discovery, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877, USA; (Z.C.); (F.H.); (D.Y.); (G.Y.); (M.S.M.)
| | - Markus Eickmann
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60596 Frankfurt am Main, Germany; (M.W.); (S.C.)
- German Center for Infection Research (DZIF), Partner Site Frankfurt am Main, 60596 Frankfurt am Main, Germany
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Branch Translational Medicine and Pharmacology, 60596 Frankfurt am Main, Germany
| | - Felix Schiele
- Biotherapeutics Discovery, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Strasse 65, 88397 Biberach an der Riss, Germany; (F.L.); (F.S.)
| | - Florian Klein
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany; (K.V.); (H.G.); (M.Z.); (C.K.); (F.K.)
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany;
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 50931 Cologne, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Hans-Meerwein-Straße 2, 35043 Marburg, Germany; (S.H.); (A.K.); (C.R.); (V.K.); (M.G.S.); (M.K.); (L.S.); (J.S.); (A.W.); (L.K.); (N.B.); (M.E.)
- German Center for Infection Research (DZIF), Partner Site Giessen-Marburg-Langen, 35043 Marburg, Germany
- Correspondence:
| |
Collapse
|
7
|
Fan K, Shen Y, Xu X, Tao L, Bao T, Li J. LncRNA-WAS and lncRNA-C8807 interact with miR-142a-3p to regulate the inflammatory response in grass carp. FISH & SHELLFISH IMMUNOLOGY 2021; 111:201-207. [PMID: 33582280 DOI: 10.1016/j.fsi.2021.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 06/12/2023]
Abstract
Septicemia of grass carp is a systemic inflammatory reaction caused by bacterial infection. More and more evidences show that long non-coding RNAs (lncRNAs) can participate in the regulation of inflammatory response. In the present study, lncRNA-WAS and lncRNA-C8807 were confirmed to be involved in the inflammatory response following infection with Aeromonas hydrophila. LncRNA-WAS and lncRNA-C8807 could interact with miR-142a-3p. LncRNA-WAS and lncRNA-C8807 interact with miR-142a-3p to effect pro-inflammatory genes and NF-κB pathway. Our results provide a theoretical basis for studying the molecular mechanism underlying the regulation of inflammation by lncRNA in grass carp.
Collapse
Affiliation(s)
- Kun Fan
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China
| | - Yubang Shen
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Xiaoyan Xu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
| | - Lizhu Tao
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China
| | - Tianjie Bao
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China
| | - Jiale Li
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
8
|
Bertrams W, Jung AL, Schmeck B. Modeling of Pneumonia and Acute Lung Injury: Bioinformatics, Systems Medicine, and Artificial Intelligence. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11689-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
9
|
Infection of pulmonary epithelial cells by clinical strains of M. tuberculosis induces alternate splicing events. Gene 2020; 750:144755. [PMID: 32380040 DOI: 10.1016/j.gene.2020.144755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 11/21/2022]
|
10
|
A MicroRNA Network Controls Legionella pneumophila Replication in Human Macrophages via LGALS8 and MX1. mBio 2020; 11:mBio.03155-19. [PMID: 32209695 PMCID: PMC7157531 DOI: 10.1128/mbio.03155-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cases of Legionella pneumophila pneumonia occur worldwide, with potentially fatal outcome. When causing human disease, Legionella injects a plethora of virulence factors to reprogram macrophages to circumvent immune defense and create a replication niche. By analyzing Legionella-induced changes in miRNA expression and genomewide chromatin modifications in primary human macrophages, we identified a cell-autonomous immune network restricting Legionella growth. This network comprises three miRNAs governing expression of the cytosolic RNA receptor DDX58/RIG-I, the tumor suppressor TP53, the antibacterial effector LGALS8, and MX1, which has been described as an antiviral factor. Our findings for the first time link TP53, LGALS8, DDX58, and MX1 in one miRNA-regulated network and integrate them into a functional node in the defense against L. pneumophila. Legionella pneumophila is an important cause of pneumonia. It invades alveolar macrophages and manipulates the immune response by interfering with signaling pathways and gene transcription to support its own replication. MicroRNAs (miRNAs) are critical posttranscriptional regulators of gene expression and are involved in defense against bacterial infections. Several pathogens have been shown to exploit the host miRNA machinery to their advantage. We therefore hypothesize that macrophage miRNAs exert positive or negative control over Legionella intracellular replication. We found significant regulation of 85 miRNAs in human macrophages upon L. pneumophila infection. Chromatin immunoprecipitation and sequencing revealed concordant changes of histone acetylation at the putative promoters. Interestingly, a trio of miRNAs (miR-125b, miR-221, and miR-579) was found to significantly affect intracellular L. pneumophila replication in a cooperative manner. Using proteome-analysis, we pinpointed this effect to a concerted downregulation of galectin-8 (LGALS8), DExD/H-box helicase 58 (DDX58), tumor protein P53 (TP53), and then MX dynamin-like GTPase 1 (MX1) by the three miRNAs. In summary, our results demonstrate a new miRNA-controlled immune network restricting Legionella replication in human macrophages.
Collapse
|
11
|
Sorting of Phagocytic Cells Infected with Legionella pneumophila. Methods Mol Biol 2019; 1921:179-189. [PMID: 30694492 DOI: 10.1007/978-1-4939-9048-1_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2023]
Abstract
The ability of Legionella pneumophila to colonize host cells and to form a replicative vacuole depends on its ability to counteract the host cell response by secreting more than 300 effectors. The host cell responds to this bacterial invasion with extensive intracellular signaling to counteract the replication of the pathogen. When studying L. pneumophila infection in vitro, only a small proportion of the cell lines or primary cells used to analyze the host response are infected; the study of such a mixed cell population leads to unprecise results. In order to study the multitude of pathogen-induced phenotypic changes occurring in the host cell, the separation of infected from uninfected cells is a top priority. Here we describe a highly efficient FACS-derived protocol to separate cells infected with a L. pneumophila strain encoding a fluorescent protein. Indeed, the highly infected, homogenous cell population obtained after sorting is the best possible starting point for the studies of infection-induced effects.
Collapse
|
12
|
Jia L, Shi Y, Wen Y, Li W, Feng J, Chen C. The roles of TNFAIP2 in cancers and infectious diseases. J Cell Mol Med 2018; 22:5188-5195. [PMID: 30145807 PMCID: PMC6201362 DOI: 10.1111/jcmm.13822] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/21/2018] [Accepted: 07/05/2018] [Indexed: 12/30/2022] Open
Abstract
TNFα‐induced protein 2 (TNFAIP2) is a primary response gene of TNFα. TNFAIP2 is highly expressed in immune cells and the urinary bladder. The expression of TNFAIP2 is regulated by multiple transcription factors and signalling pathways, including NF‐κB, KLF5 and retinoic acid. Physiologically, TNFAIP2 appears to be a multiple functional mediator not only for inflammation, angiogenesis and tunneling nanotube (TNT) formation but also as a regulator of cell proliferation and migration. The expression of TNFAIP2 is frequently abnormal in human cancers and in infectious diseases. Due to its significant functions in cell proliferation, angiogenesis, migration and invasion, TNFAIP2 could be a potential diagnostic biomarker and therapeutic target for cancer.
Collapse
Affiliation(s)
- Lin Jia
- Department of Biology, Yuxi Normal University, Yuxi, China
| | - Yundong Shi
- Department of Biology, Yuxi Normal University, Yuxi, China
| | - Yi Wen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Wei Li
- Department of Urology of the First People's Hospital of Yunnan Province, Kunming, China.,Medical College of Kunming University of Science and Technology, Kunming, China
| | - Jing Feng
- Department of Laboratory Medicine & Central Laboratory, Southern Medical University Affiliated Fengxian Hospital, Shanghai, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
13
|
Jentho E, Bodden M, Schulz C, Jung AL, Seidel K, Schmeck B, Bertrams W. microRNA-125a-3p is regulated by MyD88 in Legionella pneumophila infection and targets NTAN1. PLoS One 2017; 12:e0176204. [PMID: 28445535 PMCID: PMC5406027 DOI: 10.1371/journal.pone.0176204] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/06/2017] [Indexed: 12/01/2022] Open
Abstract
Background Legionella pneumophila (L. pneumophila) is a causative agent of severe pneumonia. It is highly adapted to intracellular replication and manipulates host cell functions like vesicle trafficking and mRNA translation to its own advantage. However, it is still unknown to what extent microRNAs (miRNAs) are involved in the Legionella-host cell interaction. Methods WT and MyD88-/- murine bone marrow-derived macrophages (BMM) were infected with L. pneumophila, the transcriptome was analyzed by high throughput qPCR array (microRNAs) and conventional qPCR (mRNAs), and mRNA-miRNA interaction was validated by luciferase assays with 3´-UTR mutations and western blot. Results L. pneumophila infection caused a pro-inflammatory reaction and significant miRNA changes in murine macrophages. In MyD88-/- cells, induction of inflammatory markers, such as Ccxl1/Kc, Il6 and miR-146a-5p was reduced. Induction of miR-125a-3p was completely abrogated in MyD88-/- cells. Target prediction analyses revealed N-terminal asparagine amidase 1 (NTAN1), a factor from the n-end rule pathway, to be a putative target of miR-125a-3p. This interaction could be confirmed by luciferase assay and western blot. Conclusion Taken together, we characterized the miRNA regulation in L. pneumophila infection with regard to MyD88 signaling and identified NTAN1 as a target of miR-125a-3p. This finding unravels a yet unknown feature of Legionella-host cell interaction, potentially relevant for new treatment options.
Collapse
Affiliation(s)
- Elisa Jentho
- Institute for Lung Research/iLung, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Malena Bodden
- Institute for Lung Research/iLung, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Christine Schulz
- Institute for Lung Research/iLung, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Anna-Lena Jung
- Institute for Lung Research/iLung, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Kerstin Seidel
- Institute for Lung Research/iLung, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| | - Bernd Schmeck
- Institute for Lung Research/iLung, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
- Department of Medicine, Pulmonary and Critical Care Medicine, University Medical Center Marburg, Philipps-University Marburg, Marburg, Germany
- * E-mail:
| | - Wilhelm Bertrams
- Institute for Lung Research/iLung, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|