1
|
Liu S, Chen L, Shang Y. CEACAM5 exacerbates asthma by inducing ferroptosis and autophagy in airway epithelial cells through the JAK/STAT6-dependent pathway. Redox Rep 2025; 30:2444755. [PMID: 39844719 PMCID: PMC11758806 DOI: 10.1080/13510002.2024.2444755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVES Asthma, a prevalent chronic disease, poses significant health threats and burdens healthcare systems. This study focused on the role of bronchial epithelial cells in asthma pathophysiology. METHODS Bioinformatics was used to identify key asthmarelated genes. An ovalbumin-sensitized mouse model and an IL-13-stimulated Beas-2B cell model were established for further investigation. RESULTS Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) was identified as a crucial gene in asthma. CEACAM5 expression was elevated in asthmatic mouse lung tissues and IL-13-stimulated Beas-2B cells, primarily in bronchial epithelial cells. CEACAM5 induced reactive oxygen species (ROS), lipid peroxidation, and ferroptosis. Interfering with CEACAM5 reduced ROS, malondialdehyde levels, and enhanced antioxidant capacity, while inhibiting iron accumulation and autophagy. Overexpression of CEACAM5 in IL-13-stimulated cells activated the JAK/STAT6 pathway, which was necessary for CEACAM5-induced autophagy, ROS accumulation, lipid peroxidation, and ferroptosis. CONCLUSION CEACAM5 promotes ferroptosis and autophagy in airway epithelial cells via the JAK/STAT6 pathway, exacerbating asthma symptoms. It represents a potential target for clinical treatment.
Collapse
Affiliation(s)
- Si Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Li Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
2
|
Safwat S, Ishak RAH, Hathout RM, Mortada ND. Bioinspired caffeic acid-laden milk protein-based nanoparticles targeting folate receptors for breast cancer treatment. Ther Deliv 2025; 16:43-61. [PMID: 39589423 PMCID: PMC11703524 DOI: 10.1080/20415990.2024.2433938] [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: 09/06/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024] Open
Abstract
AIMS Breast cancer is the second leading cause of death worldwide. Conventional chemotherapeutic therapies lack the specific targeting effect toward the cancerous cells resulting in extensive side effects. Our current study endeavors to prepare novel bioinspired folic acid-functionalized caffeic acid (CA)-loaded casein nanoparticles (CS NPs) for curbing breast cancer. METHODS CA-CS NPs were prepared by simple coacervation method followed by lyophilization. Functionalized CS NPs were achieved using folic acid as the targeting moiety. Entire comparative characterization between unconjugated and conjugated NPs were implemented in terms of size, polydispersity index, surface charge, 1H-NMR, surface morphology, in-vitro drug release, sterilization, cytotoxicity, and animal studies. RESULTS Conjugated NPs attained PS = 157.23 ± 2.64 nm, PDI = 0.309 ± 0.199, ZP = -25.53 ± 2.31 mV and IC50 = 40 ± 2.9 µg/ml. Significant reduction in the biochemical marker levels of Carcino-embryonic antigen, carbohydrate antigen 15-3, and malondialdehyde while increased superoxide dismutase levels were achieved in the tumor -induced rats treated by the conjugated NPs. Histopathological examinations showed great improvement in the mammary and necrotic regions. CONCLUSION The present work paves the road of 'back to nature' approach in designing biocompatible bioinspired conjugated nanocarriers for the diagnosis and treatment of various diseases.
Collapse
Affiliation(s)
- Sally Safwat
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Rania A. H. Ishak
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Rania M. Hathout
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nahed D. Mortada
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
3
|
Zamani MR, Šácha P. TIM3 in COVID-19; A potential hallmark? Heliyon 2024; 10:e40386. [PMID: 39759854 PMCID: PMC11700678 DOI: 10.1016/j.heliyon.2024.e40386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 01/07/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly contagious viral disease, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It can manifest as mild to severe flu-like and non-flu-like symptoms and signs, which are associated with immune dysfunction and increased mortality. The findings from COVID-19 patients imply a link between immune system abnormalities such as impaired T-cell responses or cytokine imbalances and increased risk for worse clinical outcomes, which has not been fully understood. Owing to the regulatory role of inhibitory immune checkpoints during COVID-19 infection, this review summarizes the available studies concerning the TIM3 as a relatively less characterized immune checkpoint in COVID-19 patients.
Collapse
Affiliation(s)
- Mohammad Reza Zamani
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Pavel Šácha
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
4
|
Dery KJ, Najjar SM, Beauchemin N, Shively JE, Kupiec‐Weglinski JW. Mechanism and function of CEACAM1 splice isoforms. Eur J Clin Invest 2024; 54 Suppl 2:e14350. [PMID: 39674874 PMCID: PMC11646291 DOI: 10.1111/eci.14350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/29/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Alternative splicing is a fundamental mechanism in the post-transcriptional regulation of genes. The multifunctional transmembrane glycoprotein receptor carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) undergoes extensive alternative splicing to allow for tunable functions in cell signalling, adhesion and modulation of immune and metabolic responses. Splice isoforms that differ in their ectodomain and short or long cytoplasmic tail (CEACAM1-S/CEACAM1-L) have distinct functional roles. The mechanisms that regulate CEACAM1 RNA splicing remain elusive. METHODS This narrative review summarizes the current knowledge of the mechanism and function of CEACAM1 splice isoforms. Historical perspectives address the biological significance of the glycosylated Ig domains, the variable exon 7, and phosphorylation events that dictate its signal transduction pathways. The use of small antisense molecules to target mis-spliced variable exon 7 is discussed. RESULTS The Ig variable-like N domain mediates cell adhesion and immune checkpoint inhibitory functions. Gly and Tyr residues in the transmembrane (TM) domain are essential for dimerization. Calmodulin, Calcium/Calmodulin-dependent protein kinase II delta (CamK2D), Actin and Annexin A2 are binding partners of CEACAM1-S. Homology studies of the muCEACAM1-S and huCEACAM1-S TM predict differences in their signal transduction pathways. Hypoxia-inducible factor 1-α (HIF-1-α) induces alternative splicing to produce CEACAM1-S under limited oxygen conditions. Antisense small molecules directed to exon 7 may correct faulty expression of the short and long cytoplasmic tail splicing isoforms. CONCLUSION More pre-clinical and clinical studies are needed to elucidate the precise mechanisms by which CEACAM1 RNA splicing may be exploited to develop targeted interventions towards novel therapeutic strategies.
Collapse
Affiliation(s)
- Kenneth J. Dery
- Department of SurgeryUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Sonia M. Najjar
- Department of Biomedical Sciences, Heritage College of Osteopathic MedicineOhio UniversityAthensOhioUSA
| | - Nicole Beauchemin
- Rosalind and Morris Goodman Cancer InstituteMcGill UniversityMontrealQuebecCanada
| | - John E. Shively
- Department of Theranostics and Immunology, Arthur D. Riggs Diabetes and Metabolism Research Institute, Beckman Research InstituteCity of Hope National Medical CenterDuarteCaliforniaUSA
| | | |
Collapse
|
5
|
Li Y, Mao X, Shi P, Wan Z, Yang D, Ma T, Wang B, Wang J, Wang J, Zhu R. Microbiome-host interactions in the pathogenesis of acute exacerbation of chronic obstructive pulmonary disease. Front Cell Infect Microbiol 2024; 14:1386201. [PMID: 39091676 PMCID: PMC11291260 DOI: 10.3389/fcimb.2024.1386201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/24/2024] [Indexed: 08/04/2024] Open
Abstract
Objective To explore the underlying mechanisms the airway microbiome contributes to Acute Exacerbation of Chronic Obstructive Pulmonary Disease(AECOPD). Methods We enrolled 31 AECOPD patients and 26 stable COPD patients, their sputum samples were collected for metagenomic and RNA sequencing, and then subjected to bioinformatic analyses. The expression of host genes was validated by Quantitative Real-time PCR(qPCR) using the same batch of specimens. Results Our results indicated a higher expression of Rothia mucilaginosa(p=0.015) in the AECOPD group and Haemophilus influenzae(p=0.005) in the COPD group. The Different expressed genes(DEGs) detected were significantly enriched in "type I interferon signaling pathway"(p<0.001, q=0.001) in gene function annotation, and "Cytosolic DNA-sensing pathway"(p=0.002, q=0.024), "Toll-like receptor signaling pathway"(p=0.006, q=0.045), and "TNF signaling pathway"(p=0.006, q=0.045) in KEGG enrichment analysis. qPCR amplification experiment verified that the expression of OASL and IL6 increased significantly in the AECOPD group. Conclusion Pulmonary bacteria dysbiosis may regulate the pathogenesis of AECOPD through innate immune system pathways like type I interferon signaling pathway and Toll-like receptor signaling pathway.
Collapse
Affiliation(s)
- Yao Li
- Department of Respiratory and Critical Care Medicine, Huaian Clinical College of Xuzhou Medical University, Huaian, China
| | - Xiaoyan Mao
- Department of Intensive Care Unit, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian, China
| | - Pengfei Shi
- Department of Respiratory and Critical Care Medicine, Huaian Clinical College of Xuzhou Medical University, Huaian, China
| | - Zongren Wan
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Dan Yang
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Ting Ma
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Baolan Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Jipeng Wang
- Department of Respiratory and Critical Care Medicine, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Jingjing Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rong Zhu
- Department of Respiratory and Critical Care Medicine, Huaian Clinical College of Xuzhou Medical University, Huaian, China
| |
Collapse
|
6
|
Ma RX, Wei JR, Hu YW. Characteristics of Carcinoembryonic Antigen-Related Cell Adhesion Molecules and Their Relationship to Cancer. Mol Cancer Ther 2024; 23:939-948. [PMID: 38490257 DOI: 10.1158/1535-7163.mct-23-0461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/02/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Carcinoembryonic antigen-related cell adhesion molecules (CEACAM), such as carcinoembryonic antigen (CEA) and the oncofetal glycoprotein family, are tumor markers. The CEACAMs consist of 12 different human CEACAMs and 5 different murine CEACAMs. The CEACAM family of proteins participates in multiple biological processes that include the immune response, angiogenesis, and cancer. CEACAMs play a significant role in cancer initiation and development. Increasing evidence suggests that family members may be new cancer biomarkers and targets in that CEACEAMs tend to be aberrantly expressed and therefore may have potential diagnostic and therapeutic importance. This review systematically summarizes the biogenesis, biological properties, and functions of CEACAMs, with a focus on their relationship with cancer and potential clinical application. As our knowledge of the relationships among CEACAMs and cancer increases, and as our understanding of the involved molecular mechanisms improves, new therapeutic strategies will evolve for cancer prevention and treatment of patients with cancer.
Collapse
Affiliation(s)
- Ru-Xue Ma
- Department of Cardiac Center, Guangzhou Medical University, Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Jian-Rui Wei
- Department of Cardiac Center, Guangzhou Medical University, Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Yan-Wei Hu
- Department of Laboratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
Montes de Jesus FM, Giovanella L. Unexplained increase of serum carcinoembryonic antigen: don't forget the thyroid! Clin Chem Lab Med 2023; 61:e203-e205. [PMID: 37053395 DOI: 10.1515/cclm-2023-0324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 04/02/2023] [Indexed: 04/15/2023]
Affiliation(s)
- Filipe Miguel Montes de Jesus
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Luca Giovanella
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Clinic for Nuclear Medicine, University Hospital of Zürich, Zürich, Switzerland
| |
Collapse
|
8
|
Azari F, Kennedy GT, Chang A, Bernstein E, Nadeem B, Pèlegrin A, Cailler F, Sullivan NT, Kucharczuk J, Singhal S. Glycoprotein Receptor CEACAM5-Targeted Intraoperative Molecular Imaging Tracer in Non-Small Cell Lung Cancer. Ann Thorac Surg 2023; 116:631-641. [PMID: 35644263 PMCID: PMC9701246 DOI: 10.1016/j.athoracsur.2022.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/19/2022] [Accepted: 05/09/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Intraoperative molecular imaging has emerged as a potential tool in addressing challenges faced during lung cancer surgery by localizing small lesions, ensuring negative margins, and identifying synchronous cancers. Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) glycoprotein has emerged as a potential target in fluorescent labeling of non-small cell lung cancer given the high antigen density in tumor cells and absence of expression in normal parenchyma. The goal of our study was to determine whether anti-CEACAM5 targeted near-infrared fluorochrome could be a suitable target in non-small cell lung cancer. METHODS The CEACAM5 expression was evaluated in AB-12 (known negative control), HT29 (known positive control), and H460 (non-small cell lung cancer) cell lines by polymerase chain reaction. SGM-101, a CEACAM5 antibody, coupled with a BM-104 near-infrared fluorescent tracer was evaluated with dose escalation, in vitro cellular localization, and immunofluorescence microscopy. Subsequently, in vivo validation was performed in 52 athymic nude xenografts. RESULTS Polymerase chain reaction analysis demonstrated 3000x relative expression of CEACAM5 in HT-29 cells compared with AB-12. The H460 cells showed 1000x relative expression compared with AB12 (P < .05). Both HT29 and H460 cells showed tracer internalization with signal to background ratio of 4.5 (SD 0.34) whereas there was minimal uptake by AB12 cells with signal to background ratio 1.1 (SD 0.1; P < .05). There was linear fluorescence increase with increasing tracer dosing in receptor expressing cell lines. In preclinical models, HT-29 and H460 cells lines produced near-infrared fluorescence with average tumor to background ratio of 3.89 (SD 0.25) irrespective of tumor size compared with no fluorescence by AB12 tumors (P < .05). The CEACAM5 expressing tumors had excellent dye uptake compared with AB12 tumors. CONCLUSIONS CEACAM5 serves as a possible receptor for targeted intraoperative molecular imaging resections in lung cancer. This study sets a path for evaluation of CEACAM5 targets in future clinical trials.
Collapse
Affiliation(s)
- Feredun Azari
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gregory T Kennedy
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ashley Chang
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth Bernstein
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Bilal Nadeem
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - André Pèlegrin
- SurgiMab, Montpellier, France; Montpellier Cancer Research Institute, University of Montpellier, INSERM, ICM, Montpellier, France
| | | | - Neil T Sullivan
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - John Kucharczuk
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
9
|
Su YC, Kadari M, Straw ML, Janoušková M, Jonsson S, Thofte O, Jalalvand F, Matuschek E, Sandblad L, Végvári Á, Zubarev RA, Riesbeck K. Non-typeable Haemophilus influenzae major outer membrane protein P5 contributes to bacterial membrane stability, and affects the membrane protein composition crucial for interactions with the human host. Front Cell Infect Microbiol 2023; 13:1085908. [PMID: 37305414 PMCID: PMC10250671 DOI: 10.3389/fcimb.2023.1085908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/10/2023] [Indexed: 06/13/2023] Open
Abstract
Non-typeable Haemophilus influenzae (NTHi) is a Gram-negative human pathogen that causes a wide range of airway diseases. NTHi has a plethora of mechanisms to colonize while evading the host immune system for the establishment of infection. We previously showed that the outer membrane protein P5 contributes to bacterial serum resistance by the recruitment of complement regulators. Here, we report a novel role of P5 in maintaining bacterial outer membrane (OM) integrity and protein composition important for NTHi-host interactions. In silico analysis revealed a peptidoglycan-binding motif at the periplasmic C-terminal domain (CTD) of P5. In a peptidoglycan-binding assay, the CTD of P5 (P5CTD) formed a complex with peptidoglycan. Protein profiling analysis revealed that deletion of CTD or the entire P5 changed the membrane protein composition of the strains NTHi 3655Δp5CTD and NTHi 3655Δp5, respectively. Relative abundance of several membrane-associated virulence factors that are crucial for adherence to the airway mucosa, and serum resistance were altered. This was also supported by similar attenuated pathogenic phenotypes observed in both NTHi 3655Δp5 CTD and NTHi 3655Δp5. We found (i) a decreased adherence to airway epithelial cells and fibronectin, (ii) increased complement-mediated killing, and (iii) increased sensitivity to the β-lactam antibiotics in both mutants compared to NTHi 3655 wild-type. These mutants were also more sensitive to lysis at hyperosmotic conditions and hypervesiculated compared to the parent wild-type bacteria. In conclusion, our results suggest that P5 is important for bacterial OM stability, which ultimately affects the membrane proteome and NTHi pathogenesis.
Collapse
Affiliation(s)
- Yu-Ching Su
- Department of Translational Medicine, Clinical Microbiology, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Mahendar Kadari
- Department of Translational Medicine, Clinical Microbiology, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Megan L. Straw
- Department of Translational Medicine, Clinical Microbiology, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Martina Janoušková
- Department of Translational Medicine, Clinical Microbiology, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Sandra Jonsson
- Department of Translational Medicine, Clinical Microbiology, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Oskar Thofte
- Department of Translational Medicine, Clinical Microbiology, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Farshid Jalalvand
- Department of Translational Medicine, Clinical Microbiology, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Erika Matuschek
- European Committee on Antimicrobial Susceptibility Testing (EUCAST) Development Laboratory, c/o Clinical Microbiology, Central Hospital, Växjö, Sweden
| | - Linda Sandblad
- Department of Chemistry and The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry & Biophysics (MBB), Proteomics Biomedicum, Karolinska Institute, Stockholm, Sweden
| | - Roman A. Zubarev
- Division of Chemistry I, Department of Medical Biochemistry & Biophysics (MBB), Proteomics Biomedicum, Karolinska Institute, Stockholm, Sweden
| | - Kristian Riesbeck
- Department of Translational Medicine, Clinical Microbiology, Faculty of Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
10
|
Herrera-Luis E, Forno E, Celedón JC, Pino-Yanes M. Asthma Exacerbations: The Genes Behind the Scenes. J Investig Allergol Clin Immunol 2023; 33:76-94. [PMID: 36420738 PMCID: PMC10638677 DOI: 10.18176/jiaci.0878] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The clinical and socioeconomic burden of asthma exacerbations (AEs) constitutes a major public health problem. In the last 4 years, there has been an increase in ethnic diversity in candidate-gene and genome-wide association studies of AEs, which in the latter case led to the identification of novel genes and underlying pathobiological processes. Pharmacogenomics, admixture mapping analyses, and the combination of multiple "omics" layers have helped to prioritize genomic regions of interest and/or facilitated our understanding of the functional consequences of genetic variation. Nevertheless, the field still lags behind the genomics of asthma, where a vast compendium of genetic approaches has been used (eg, gene-environment nteractions, next-generation sequencing, and polygenic risk scores). Furthermore, the roles of the DNA methylome and histone modifications in AEs have received little attention, and microRNA findings remain to be validated in independent studies. Likewise, the most recent transcriptomic studies highlight the importance of the host-airway microbiome interaction in the modulation of risk of AEs. Leveraging -omics and deep-phenotyping data from subtypes or homogenous subgroups of patients will be crucial if we are to overcome the inherent heterogeneity of AEs, boost the identification of potential therapeutic targets, and implement precision medicine approaches to AEs in clinical practice.
Collapse
Affiliation(s)
- E Herrera-Luis
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain
| | - E Forno
- Division of Pediatric Pulmonary Medicine, UPMC Children´s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - J C Celedón
- Division of Pediatric Pulmonary Medicine, UPMC Children´s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - M Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain 4 Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), La Laguna, Tenerife, Spain
| |
Collapse
|
11
|
Dissanayake E, Brockman-Schneider RA, Stubbendieck RM, Helling BA, Zhang Z, Bochkov YA, Kirkham C, Murphy TF, Ober C, Currie CR, Gern JE. Rhinovirus increases Moraxella catarrhalis adhesion to the respiratory epithelium. Front Cell Infect Microbiol 2023; 12:1060748. [PMID: 36733852 PMCID: PMC9886879 DOI: 10.3389/fcimb.2022.1060748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/28/2022] [Indexed: 01/18/2023] Open
Abstract
Rhinovirus causes many types of respiratory illnesses, ranging from minor colds to exacerbations of asthma. Moraxella catarrhalis is an opportunistic pathogen that is increased in abundance during rhinovirus illnesses and asthma exacerbations and is associated with increased severity of illness through mechanisms that are ill-defined. We used a co-infection model of human airway epithelium differentiated at the air-liquid interface to test the hypothesis that rhinovirus infection promotes M. catarrhalis adhesion and survival on the respiratory epithelium. Initial experiments showed that infection with M. catarrhalis alone did not damage the epithelium or induce cytokine production, but increased trans-epithelial electrical resistance, indicative of increased barrier function. In a co-infection model, infection with the more virulent rhinovirus-A and rhinovirus-C, but not the less virulent rhinovirus-B types, increased cell-associated M. catarrhalis. Immunofluorescent staining demonstrated that M. catarrhalis adhered to rhinovirus-infected ciliated epithelial cells and infected cells being extruded from the epithelium. Rhinovirus induced pronounced changes in gene expression and secretion of inflammatory cytokines. In contrast, M. catarrhalis caused minimal effects and did not enhance RV-induced responses. Our results indicate that rhinovirus-A or C infection increases M. catarrhalis survival and cell association while M. catarrhalis infection alone does not cause cytopathology or epithelial inflammation. Our findings suggest that rhinovirus and M. catarrhalis co-infection could promote epithelial damage and more severe illness by amplifying leukocyte inflammatory responses at the epithelial surface.
Collapse
Affiliation(s)
- Eishika Dissanayake
- Department of Pediatrics, University of Wisconsin – Madison, Madison, WI, United States
| | | | - Reed M. Stubbendieck
- Department of Bacteriology, University of Wisconsin – Madison, Madison, WI, United States
| | - Britney A. Helling
- Department of Human Genetics, University of Chicago, Chicago, IL, United States
| | - Zhumin Zhang
- Department of Biostatistics and Medical Informatics, University of Wisconsin – Madison, Madison, WI, United States
| | - Yury A. Bochkov
- Department of Pediatrics, University of Wisconsin – Madison, Madison, WI, United States
| | - Charmaine Kirkham
- Clinical and Translational Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Timothy F. Murphy
- Clinical and Translational Research Center, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, United States
| | - Cameron R. Currie
- Department of Bacteriology, University of Wisconsin – Madison, Madison, WI, United States
- Michael G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - James E. Gern
- Department of Pediatrics, University of Wisconsin – Madison, Madison, WI, United States
| |
Collapse
|
12
|
Azari F, Meijer RPJ, Kennedy GT, Hanna A, Chang A, Nadeem B, Din A, Pèlegrin A, Framery B, Cailler F, Sullivan NT, Kucharczuk J, Martin LW, Vahrmeijer AL, Singhal S. Carcinoembryonic Antigen-Related Cell Adhesion Molecule Type 5 Receptor-Targeted Fluorescent Intraoperative Molecular Imaging Tracer for Lung Cancer: A Nonrandomized Controlled Trial. JAMA Netw Open 2023; 6:e2252885. [PMID: 36705924 PMCID: PMC10292762 DOI: 10.1001/jamanetworkopen.2022.52885] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Importance Localization of subcentimeter ground glass opacities during minimally invasive thoracoscopic lung cancer resections is a significant challenge in thoracic oncology. Intraoperative molecular imaging has emerged as a potential solution, but the availability of suitable fluorescence agents is a limiting factor. Objective To evaluate the suitability of SGM-101, a carcinoembryonic antigen-related cell adhesion molecule type 5 (CEACAM5) receptor-targeted near-infrared fluorochrome, for molecular imaging-guided lung cancer resections, because glycoprotein is expressed in more than 80% of adenocarcinomas. Design, Setting, and Participants For this nonrandomized, proof-of-principal, phase 1 controlled trial, patients were divided into 2 groups between August 1, 2020, and January 31, 2022. Patients with known CEACAM5-positive gastrointestinal tumors suggestive of lung metastasis were selected as proof-of-principle positive controls. The investigative group included patients with lung nodules suggestive of primary lung malignant neoplasms. Patients 18 years or older without significant comorbidities that precluded surgical exploration with suspicious pulmonary nodules requiring surgical biopsy were included in the study. Interventions SGM-101 (10 mg) was infused up to 5 days before index operation, and pulmonary nodules were imaged using a near-infrared camera system with a dedicated thoracoscope. Main Outcomes and Measures SGM-101 localization to pulmonary nodules and its correlation with CEACAM5 glycoprotein expression by the tumor as quantified by tumor and normal pulmonary parenchymal fluorescence. Results Ten patients (5 per group; 5 male and 5 female; median [IQR] age, 66 [58-69] years) with 14 total lesions (median [range] lesion size, 0.91 [0.90-2.00] cm) were enrolled in the study. In the control group of 4 patients (1 patient did not undergo surgical resection because of abnormal preoperative cardiac clearance findings that were not deemed related to SGM-101 infusion), the mean (SD) lesion size was 1.33 (0.48) cm, 2 patients had elevated serum CEA markers, and 2 patients had normal serum CEA levels. Of the 4 patients who underwent surgical intervention, those with 2+ and 3+ tissue CEACAM5 expression had excellent tumor fluorescence, with a mean (SD) tumor to background ratio of 3.11 (0.45). In the patient cohort, the mean (SD) lesion size was 0.68 (0.22) cm, and no elevations in serum CEA levels were found. Lack of SGM-101 fluorescence was associated with benign lesions and with lack of CEACAM5 staining. Conclusions and Relevance This in-human proof-of-principle nonrandomized controlled trial demonstrated SGM-101 localization to CEACAM5-positive tumors with the detection of real-time near-infrared fluorescence in situ, ex vivo, and by immunofluorescence microscopy. These findings suggest that SGM-101 is a safe, receptor-specific, and feasible intraoperative molecular imaging fluorochrome that should be further evaluated in randomized clinical trials. Trial Registration ClinicalTrials.gov identifier: NCT04315467.
Collapse
Affiliation(s)
- Feredun Azari
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia
| | - Ruben P J Meijer
- Centre for Human Drug Research, Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Gregory T Kennedy
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia
| | - Andrew Hanna
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Ashley Chang
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia
| | - Bilal Nadeem
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia
| | - Azra Din
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia
| | - André Pèlegrin
- SurgiMab, Montpellier, France
- Institute of Cancer Research of Montpellier, University of Montpellier, Montpellier, France
| | | | | | - Neil T Sullivan
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia
| | - John Kucharczuk
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia
| | - Linda W Martin
- Department of Thoracic Surgery, University of Virginia School of Medicine, Charlottesville
| | - Alexander L Vahrmeijer
- Centre for Human Drug Research, Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Sunil Singhal
- Department of Thoracic Surgery, University of Pennsylvania, Philadelphia
| |
Collapse
|
13
|
Alhetheel A, Albarrag A, Shakoor Z, Somily A, Barry M, Altalhi H, Bakhrebah M, Nassar M, Alfageeh M, Assiri A, Alfaraj S, Memish ZA. Differential expression of carcinoembryonic antigen-related cell adhesion molecule-5 (CEACAM5) and dipeptidyl peptidase-4 (DPP4) with detection of Middle East respiratory syndrome-coronavirus in peripheral blood. J Infect Public Health 2022; 15:1315-1320. [PMID: 36279687 PMCID: PMC9576204 DOI: 10.1016/j.jiph.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/13/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Middle East respiratory syndrome-coronavirus (MERS-CoV) utilizes CD26 (dipeptidyl peptidase-4) and CD66e or CEACAM5 (carcinoembryonic antigen-related cell adhesion molecule 5) receptors for cell infection. Peripheral blood mononuclear cells (PBMCs) play a critical role in mounting adaptive immune response against the virus. This study was performed to assess the expression of CD26 and CD66e on PBMCs and their susceptibility to MERS-CoV infection. METHODS Surface expression of CD26 and CD66e receptors on PBMCs from MERS-CoV patients (n = 20) and healthy controls (n = 20) was assessed by flow cytometry and the soluble forms were determined by enzyme-linked immunosorbent assay (ELISA). MERS-CoV UpE and Orf1a genes in PBMCs were detected by using Altona diagnostics reverse transcription polymerase chain reaction (RT-PCR) kit. RESULTS Mean fluorescent intensity (MFI) of CD66e was significantly higher on CD4 + lymphocytes (462.4 ± 64.35 vs 325.1 ± 19.69; p < 0.05) and CD8 + lymphocytes (533.8 ± 55.32 vs 392.4 ± 37.73; p < 0.04) from patients with MERS-CoV infection compared to the normal controls. No difference in MFI for CD66e was observed on monocytes (381.8 ± 40.34 vs 266.8 ± 20.6; p = 0.3) between the patients and controls. Soluble form of CD66e among MERS-CoV patients was also higher than the normal controls (mean= 338.7 ± 58.75 vs 160.7 ± 29.49 ng/mL; p < 0.01). Surface expression of CD26 on PBMCs and its soluble form were no different between the groups. MERS-CoV was detected by RT-PCR in 16/20 (80%) patients from whole blood, among them 8 patients were tested in PBMCs, 4/8 (50%) patients were positive. CONCLUSION Increased expression levels of CD66e (CEACAM5) may contribute to increased susceptibility of PBMCs to MERS-CoV infection and disease progression.
Collapse
Affiliation(s)
- Abdulkarim Alhetheel
- King Khalid University Hospital, Riyadh, Saudi Arabia; Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| | - Ahmed Albarrag
- King Khalid University Hospital, Riyadh, Saudi Arabia; Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Zahid Shakoor
- King Khalid University Hospital, Riyadh, Saudi Arabia; Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Ali Somily
- King Khalid University Hospital, Riyadh, Saudi Arabia; Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mazin Barry
- King Khalid University Hospital, Riyadh, Saudi Arabia; Department of Infectious Diseases, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Hifa Altalhi
- King Khalid University Hospital, Riyadh, Saudi Arabia
| | | | - Majed Nassar
- King Abdulaziz city for Science and Technology, Riyadh, Saudi Arabia
| | - Mohamed Alfageeh
- King Abdulaziz city for Science and Technology, Riyadh, Saudi Arabia
| | - Ayed Assiri
- Critical Care Unit, Prince Mohammed Bin Abdulaziz Hospital, Ministry of Health, Riyadh, Saudi Arabia
| | - Sarah Alfaraj
- Corona Center, Prince Mohammed Bin Abdulaziz Hospital, Ministry of Health, Riyadh, Saudi Arabia
| | - Ziad A Memish
- Research and Innovation Center, King Saud Medical City, Ministry of Health, Riyadh, Saudi Arabia; College of Medicine, Alfaisal University, Riyadh, Saudi Arabia; Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| |
Collapse
|
14
|
Davodabadi F, Sarhadi M, Arabpour J, Sargazi S, Rahdar A, Díez-Pascual AM. Breast cancer vaccines: New insights into immunomodulatory and nano-therapeutic approaches. J Control Release 2022; 349:844-875. [PMID: 35908621 DOI: 10.1016/j.jconrel.2022.07.036] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022]
Abstract
Breast cancer (BC) is known to be a highly heterogeneous disease that is clinically subdivided into four primary molecular subtypes, each having distinct morphology and clinical implications. These subtypes are principally defined by hormone receptors and other proteins involved (or not involved) in BC development. BC therapeutic vaccines [including peptide-based vaccines, protein-based vaccines, nucleic acid-based vaccines (DNA/RNA vaccines), bacterial/viral-based vaccines, and different immune cell-based vaccines] have emerged as an appealing class of cancer immunotherapeutics when used alone or combined with other immunotherapies. Employing the immune system to eliminate BC cells is a novel therapeutic modality. The benefit of active immunotherapies is that they develop protection against neoplastic tissue and readjust the immune system to an anti-tumor monitoring state. Such immunovaccines have not yet shown effectiveness for BC treatment in clinical trials. In recent years, nanomedicines have opened new windows to increase the effectiveness of vaccinations to treat BC. In this context, some nanoplatforms have been designed to efficiently deliver molecular, cellular, or subcellular vaccines to BC cells, increasing the efficacy and persistence of anti-tumor immunity while minimizing undesirable side effects. Immunostimulatory nano-adjuvants, liposomal-based vaccines, polymeric vaccines, virus-like particles, lipid/calcium/phosphate nanoparticles, chitosan-derived nanostructures, porous silicon microparticles, and selenium nanoparticles are among the newly designed nanostructures that have been used to facilitate antigen internalization and presentation by antigen-presenting cells, increase antigen stability, enhance vaccine antigenicity and remedial effectivity, promote antigen escape from the endosome, improve cytotoxic T lymphocyte responses, and produce humoral immune responses in BC cells. Here, we summarized the existing subtypes of BC and shed light on immunomodulatory and nano-therapeutic strategies for BC vaccination. Finally, we reviewed ongoing clinical trials on BC vaccination and highlighted near-term opportunities for moving forward.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran.
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran.
| | - Ana M Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
15
|
Paris T, Yatime L. [CEACAMs as anchoring platforms for pathogens on mucosal epithelia]. Med Sci (Paris) 2022; 38:650-653. [PMID: 36094234 DOI: 10.1051/medsci/2022097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Théo Paris
- Laboratoire des interactions hôte-pathogène, UMR5235, Université de Montpellier, CNRS, Inserm, Montpellier, France
| | - Laure Yatime
- Laboratoire des interactions hôte-pathogène, UMR5235, Université de Montpellier, CNRS, Inserm, Montpellier, France
| |
Collapse
|
16
|
Thomson NC. Frequent exacerbators in severe asthma: Focus on clinical and transcriptional factors. Clin Transl Med 2022; 12:e860. [PMID: 35538891 PMCID: PMC9091991 DOI: 10.1002/ctm2.860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 04/17/2022] [Accepted: 04/19/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Neil C Thomson
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, Scotland, UK
| |
Collapse
|
17
|
Hoda U, Pavlidis S, Bansal AT, Takahashi K, Hu S, Ng Kee Kwong F, Rossios C, Sun K, Bhavsar P, Loza M, Baribaud F, Chanez P, Fowler SJ, Horvath I, Montuschi P, Singer F, Musial J, Dahlen B, Krug N, Sandstrom T, Shaw DE, Lutter R, Fleming LJ, Howarth PH, Caruso M, Sousa AR, Corfield J, Auffray C, De Meulder B, Lefaudeux D, Dahlen SE, Djukanovic R, Sterk PJ, Guo Y, Adcock IM, Chung KF. Clinical and transcriptomic features of persistent exacerbation-prone severe asthma in U-BIOPRED cohort. Clin Transl Med 2022; 12:e816. [PMID: 35474304 PMCID: PMC9043117 DOI: 10.1002/ctm2.816] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 01/28/2023] Open
Abstract
Background Exacerbation‐prone asthma is a feature of severe disease. However, the basis for its persistency remains unclear. Objectives To determine the clinical and transcriptomic features of frequent exacerbators (FEs) and persistent FEs (PFEs) in the U‐BIOPRED cohort. Methods We compared features of FE (≥2 exacerbations in past year) to infrequent exacerbators (IE, <2 exacerbations) and of PFE with repeat ≥2 exacerbations during the following year to persistent IE (PIE). Transcriptomic data in blood, bronchial and nasal epithelial brushings, bronchial biopsies and sputum cells were analysed by gene set variation analysis for 103 gene signatures. Results Of 317 patients, 62.4% had FE, of whom 63.6% had PFE, while 37.6% had IE, of whom 61.3% had PIE. Using multivariate analysis, FE was associated with short‐acting beta‐agonist use, sinusitis and daily oral corticosteroid use, while PFE was associated with eczema, short‐acting beta‐agonist use and asthma control index. CEA cell adhesion molecule 5 (CEACAM5) was the only differentially expressed transcript in bronchial biopsies between PE and IE. There were no differentially expressed genes in the other four compartments. There were higher expression scores for type 2, T‐helper type‐17 and type 1 pathway signatures together with those associated with viral infections in bronchial biopsies from FE compared to IE, while there were higher expression scores of type 2, type 1 and steroid insensitivity pathway signatures in bronchial biopsies of PFE compared to PIE. Conclusion The FE group and its PFE subgroup are associated with poor asthma control while expressing higher type 1 and type 2 activation pathways compared to IE and PIE, respectively.
Collapse
Affiliation(s)
- Uruj Hoda
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Stelios Pavlidis
- Department of Computing & Data Science Institute, Imperial College London
| | | | - Kentaro Takahashi
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK.,Research Centre for Allergy and Clinical Immunology, Asahi General Hospital, Asahi, Japan
| | | | - Francois Ng Kee Kwong
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Christos Rossios
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | | | - Pankaj Bhavsar
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Matthew Loza
- Janssen Research and Development, High Wycombe, Buckinghamshire, UK
| | | | - Pascal Chanez
- Assistance Publique des Hôpitaux de Marseille, Clinique des Bronches, Allergies et Sommeil, Aix Marseille Université, Marseille, France
| | - Stephen J Fowler
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, and NIHR Biomedical Research Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Ildiko Horvath
- Department of Pulmonology, Semmelweis University, Budapest, Hungary
| | | | - Florian Singer
- Department of Respiratory Medicine, University Children's Hospital Zurich and Childhood Research Center, Zurich, and Department of Paediatrics, Inselspital, University of Bern, Switzerland
| | - Jacek Musial
- Department of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Barbro Dahlen
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Norbert Krug
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Thomas Sandstrom
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | | | - Rene Lutter
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Louise J Fleming
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Peter H Howarth
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Massimo Caruso
- Department of Biochemical and Biotechnological Medicine, University of Catania, Catania, Italy
| | - Ana R Sousa
- Respiratory Therapeutic Unit, GSK, Stockley Park, UK
| | - Julie Corfield
- AstraZeneca R&D, Molndal, Sweden, and Areteva R&D, Nottingham, UK
| | - Charles Auffray
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Lyon, France
| | - Bertrand De Meulder
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Lyon, France
| | - Diane Lefaudeux
- European Institute for Systems Biology and Medicine, CNRS-ENS-UCBL-INSERM, Lyon, France
| | - Sven-Erik Dahlen
- Centre for Allergy Research, Karolinska Institutet, Stockholm, Sweden
| | - Ratko Djukanovic
- NIHR Southampton Biomedical Research Centre, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, UK
| | - Peter J Sterk
- Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, and Biomedical Research Unit, Royal Brompton and Harefield NHS Trust, London, UK
| | | |
Collapse
|
18
|
Gunn AH, Tashie C, Wolf S, Troy JD, Zafar Y. Tumor marker response to SARS-CoV-2 infection among patients with cancer. Cancer Med 2022; 11:2865-2872. [PMID: 35289488 PMCID: PMC9110907 DOI: 10.1002/cam4.4646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 11/10/2022] Open
Abstract
Background Inflammatory responses from benign conditions can cause non‐cancer‐related elevations in tumor markers. The severe acute respiratory coronavirus 2 (SARS‐CoV‐2) induces a distinct viral inflammatory response, resulting in coronavirus disease 2019 (COVID‐19). Clinical data suggest carcinoembryonic antigen (CEA), carbohydrate antigen 19–9 (CA 19–9), and cancer antigen 125 (CA 125) levels might rise in patients with COVID‐19. However, available data excludes cancer patients, so little is known about the effect of COVID‐19 on tumor markers among cancer patients. Methods We conducted a case series and identified patients with a positive SARS‐CoV‐2 PCR test, diagnosis of a solid tumor malignancy, and a CEA, CA 19–9, CA 125, or CA 27–29 laboratory test. Cancer patients with documented COVID‐19 infection and at least one pre‐ and two post‐infection tumor marker measurements were included. We abstracted the electronic health record for demographics, cancer diagnosis, treatment, evidence of cancer progression, date and severity of COVID‐19 infection, and tumor marker values. Results Seven patients were identified with a temporary elevation of tumor marker values during the post‐COVID‐19 period. Elevation in tumor marker occurred within 56 days of COVID‐19 infection for all patients. Tumor markers subsequently decreased at the second time point in the post‐infectious period among all patients. Conclusion We report temporary elevations of cancer tumor markers in the period surrounding COVID‐19 infection. To our knowledge this is the first report of this phenomenon in cancer patients and has implications for clinical management and future research.
Collapse
Affiliation(s)
| | | | - Steven Wolf
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Jesse D Troy
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, USA
| | - Yousuf Zafar
- Duke University School of Medicine, Durham, North Carolina, USA.,Duke Cancer Institute, Durham, North Carolina, USA
| |
Collapse
|
19
|
Klaile E, Prada Salcedo JP, Klassert TE, Besemer M, Bothe AK, Durotin A, Müller MM, Schmitt V, Luther CH, Dittrich M, Singer BB, Dandekar T, Slevogt H. Antibody ligation of CEACAM1, CEACAM3, and CEACAM6, differentially enhance the cytokine release of human neutrophils in responses to Candida albicans. Cell Immunol 2021; 371:104459. [PMID: 34847408 DOI: 10.1016/j.cellimm.2021.104459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 11/30/2022]
Abstract
Invasive candidiasis is a healthcare-associated fungal infection with a high mortality rate. Neutrophils, the first line of defense during fungal infections, express the immunoregulatory Candida albicans receptors CEACAM1, CEACAM3, and CEACAM6. We analyzed the effects of specific antibodies on C. albicans-induced neutrophil responses. CEACAM6 ligation by 1H7-4B and to some extent CEACAM1 ligation by B3-17, but not CEACAM3 ligation by 308/3-3, resulted in the immediate release of stored CXCL8 and altered transcriptional responses of the C. albicans-stimulated neutrophils. Integrated network analyses and dynamic simulations of signaling cascades predicted alterations in apoptosis and cytokine secretion. We verified that CEACAM6 ligation enhanced Candida-induced neutrophil apoptosis and increased long-term IL-1β/IL-6 release in responses to C. albicans. CEACAM3 ligation, but not CEACAM1 ligation, increased the long-term release of pro-inflammatory IL-1β/IL-6. Taken together, we demonstrated for the first time that ligation of CEACAM receptors differentially affects the regulation of C. albicans-induced immune functions in human neutrophils.
Collapse
Affiliation(s)
- Esther Klaile
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Juan P Prada Salcedo
- Dept. of Bioinformatics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany.
| | - Tilman E Klassert
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Matthias Besemer
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Anne-Katrin Bothe
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Adrian Durotin
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Mario M Müller
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| | - Verena Schmitt
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany.
| | - Christian H Luther
- Dept. of Bioinformatics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany.
| | - Marcus Dittrich
- Dept. of Bioinformatics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany; Dept. of Human Genetics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany.
| | - Bernhard B Singer
- Institute of Anatomy, University Hospital, University Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany.
| | - Thomas Dandekar
- Dept. of Bioinformatics, University of Würzburg, Biocenter/Am Hubland, 97074 Würzburg, Germany.
| | - Hortense Slevogt
- ZIK Septomics, University Hospital Jena, Albert-Einstein-Straße 10, 07749 Jena, Germany.
| |
Collapse
|
20
|
Kowalewski J, Paris T, Gonzalez C, Lelièvre E, Castaño Valencia L, Boutrois M, Augier C, Lutfalla G, Yatime L. Characterization of a member of the CEACAM protein family as a novel marker of proton pump-rich ionocytes on the zebrafish epidermis. PLoS One 2021; 16:e0254533. [PMID: 34252160 PMCID: PMC8274849 DOI: 10.1371/journal.pone.0254533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 06/29/2021] [Indexed: 01/04/2023] Open
Abstract
In humans, several members of the CEACAM receptor family have been shown to interact with intestinal pathogens in an inflammatory context. While CEACAMs have long been thought to be only present in mammals, recent studies have identified ceacam genes in other vertebrates, including teleosts. The function of these related genes remains however largely unknown. To gain insight into the function of CEACAM proteins in fish, we undertook the study of a putative member of the family, CEACAMz1, identified in Danio rerio. Sequence analysis of the ceacamz1 gene product predicted a GPI-anchored extracellular protein containing eleven immunoglobulin domains but revealed no evident orthology with human CEACAMs. Using a combination of RT-PCR analyses and in situ hybridization experiments, as well as a fluorescent reporter line, we showed that CEACAMz1 is first expressed in discrete cells on the ventral skin of zebrafish larvae and later on in the developing gills. This distribution remains constant until juvenile stage is reached, at which point CEACAMz1 is almost exclusively expressed in gills. We further observed that at late larval stages, CEACAMz1-expressing cells mostly localize on the afferent side of the branchial filaments and possibly in the inter-lamellar space. Using immunolabelling and 3D-reconstructions, we showed that CEACAMz1 is expressed in cells from the uppermost layer of skin epidermis. These cells are embedded within the keratinocytes pavement and we unambiguously identified them as proton-pump rich ionocytes (HR cells). As the expression of ceacamz1 is turned on concomitantly to that of other known markers of HR cells, we propose that ceacamz1 may serve as a novel marker of mature HR cells from the zebrafish epidermis.
Collapse
Affiliation(s)
- Julien Kowalewski
- Laboratory of Pathogen-Host Interactions (LPHI), UMR5235, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Théo Paris
- Laboratory of Pathogen-Host Interactions (LPHI), UMR5235, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Catherine Gonzalez
- Laboratory of Pathogen-Host Interactions (LPHI), UMR5235, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Etienne Lelièvre
- Laboratory of Pathogen-Host Interactions (LPHI), UMR5235, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Lina Castaño Valencia
- Laboratory of Pathogen-Host Interactions (LPHI), UMR5235, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Morgan Boutrois
- Laboratory of Pathogen-Host Interactions (LPHI), UMR5235, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Camille Augier
- Laboratory of Pathogen-Host Interactions (LPHI), UMR5235, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Georges Lutfalla
- Laboratory of Pathogen-Host Interactions (LPHI), UMR5235, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Laure Yatime
- Laboratory of Pathogen-Host Interactions (LPHI), UMR5235, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
21
|
Huang R, Meng T, Zha Q, Cheng K, Zhou X, Zheng J, Zhang D, Liu R. The predicting roles of carcinoembryonic antigen and its underlying mechanism in the progression of coronavirus disease 2019. Crit Care 2021; 25:234. [PMID: 34217339 PMCID: PMC8254455 DOI: 10.1186/s13054-021-03661-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 06/29/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) has induced a worldwide epidemiological event with a high infectivity and mortality. However, the predicting biomarkers and their potential mechanism in the progression of COVID-19 are not well known. OBJECTIVE The aim of this study is to identify the candidate predictors of COVID-19 and investigate their underlying mechanism. METHODS The retrospective study was conducted to identify the potential laboratory indicators with prognostic values of COVID-19 disease. Then, the prognostic nomogram was constructed to predict the overall survival of COVID-19 patients. Additionally, the scRNA-seq data of BALF and PBMCs from COVID-19 patients were downloaded to investigate the underlying mechanism of the most important prognostic indicators in lungs and peripherals, respectively. RESULTS In total, 304 hospitalized adult COVID-19 patients in Wuhan Jinyintan Hospital were included in the retrospective study. CEA was the only laboratory indicator with significant difference in the univariate (P < 0.001) and multivariate analysis (P = 0.020). The scRNA-seq data of BALF and PBMCs from COVID-19 patients were downloaded to investigate the underlying mechanism of CEA in lungs and peripherals, respectively. The results revealed the potential roles of CEA were significantly distributed in type II pneumocytes of BALF and developing neutrophils of PBMCs, participating in the progression of COVID-19 by regulating the cell-cell communication. CONCLUSION This study identifies the prognostic roles of CEA in COVID-19 patients and implies the potential roles of CEACAM8-CEACAM6 in the progression of COVID-19 by regulating the cell-cell communication of developing neutrophils and type II pneumocyte.
Collapse
Affiliation(s)
- Runzhi Huang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Shanghai, 200065, China
| | - Tong Meng
- Shanghai General Hospital, 100 Haining Road, Shanghai, 200080, China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | - Qiongfang Zha
- Department of Respiratory and Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127, China
| | - Kebin Cheng
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Xin Zhou
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, 100 Haining Road, Shanghai, 200080, China
| | - Junhua Zheng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Yanchang Road, Shanghai, 200072, China
| | | | - Ruilin Liu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| |
Collapse
|
22
|
Wronski S, Beinke S, Obernolte H, Belyaev NN, Saunders KA, Lennon MG, Schaudien D, Braubach P, Jonigk D, Warnecke G, Zardo P, Fieguth HG, Wilkens L, Braun A, Hessel EM, Sewald K. Rhinovirus-induced Human Lung Tissue Responses Mimic COPD and Asthma Gene Signatures. Am J Respir Cell Mol Biol 2021; 65:544-554. [PMID: 34181859 PMCID: PMC8641849 DOI: 10.1165/rcmb.2020-0337oc] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Human rhinovirus (RV) is a major risk factor for chronic obstructive pulmonary disease (COPD) and asthma exacerbations. The exploration of RV pathogenesis has been hampered by a lack of disease-relevant model systems. We performed a detailed characterization of host responses to RV infection in human lung tissue ex vivo and investigated whether these responses are disease relevant for patients with COPD and asthma. In addition, impact of the viral replication inhibitor rupintrivir was evaluated. Human precision-cut lung slices (PCLS) were infected with RV1B with or without rupintrivir. At Days 1 and 3 after infection, RV tissue localization, tissue viability, and viral load were determined. To characterize host responses to infection, mediator and whole genome analyses were performed. RV successfully replicated in PCLS airway epithelial cells and induced both antiviral and proinflammatory cytokines such as IFNα2a, CXCL10, CXCL11, IFN-γ, TNFα, and CCL5. Genomic analyses revealed that RV not only induced antiviral immune responses but also triggered changes in epithelial cell–associated pathways. Strikingly, the RV response in PCLS was reflective of gene expression changes described in patients with COPD and asthma. Although RV-induced host immune responses were abrogated by rupintrivir, RV-triggered epithelial processes were largely refractory to antiviral treatment. Detailed analysis of RV-infected human PCLS and comparison with gene signatures of patients with COPD and asthma revealed that the human RV PCLS model represents disease-relevant biological mechanisms that can be partially inhibited by a well-known antiviral compound and provide an outstanding opportunity to evaluate novel therapeutics.
Collapse
Affiliation(s)
- Sabine Wronski
- Fraunhofer Institute for Toxicology and Experimental Medicine, Member of Fraunhofer international Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany.,Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany;
| | - Soren Beinke
- Research and Development, GlaxoSmithKline, Stevenage, United Kingdom of Great Britain and Northern Ireland
| | - Helena Obernolte
- Fraunhofer Institute for Toxicology and Experimental Medicine, Member of Fraunhofer international Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany.,Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Nikolai N Belyaev
- Research and Development, GlaxoSmithKline, Stevenage, United Kingdom of Great Britain and Northern Ireland
| | - Ken A Saunders
- Research and Development, GlaxoSmithKline, Stevenage, United Kingdom of Great Britain and Northern Ireland
| | - Mark G Lennon
- Research and Development, GlaxoSmithKline, Stevenage, United Kingdom of Great Britain and Northern Ireland
| | - Dirk Schaudien
- Fraunhofer Institute for Toxicology and Experimental Medicine, Member of Fraunhofer international Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany.,Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Dsease (BREATH), Hannover, Germany
| | - Peter Braubach
- Hannover Medical School, 9177, Department of Pathology, Hannover, Germany.,Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Danny Jonigk
- Hannover Medical School, 9177, Department of Pathology, Hannover, Niedersachsen, Germany.,Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Gregor Warnecke
- Hannover Medical School, 9177, Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover, Germany.,Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Patrick Zardo
- Hannover Medical School, 9177, Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover, Germany
| | | | | | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine, Member of Fraunhofer international Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany.,Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | - Edith M Hessel
- Research and Development, GlaxoSmithKline, Stevenage, United Kingdom of Great Britain and Northern Ireland
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine, Member of Fraunhofer international Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany.,Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease (BREATH), Hannover, Germany
| | | |
Collapse
|
23
|
The Immunomodulatory CEA Cell Adhesion Molecule 6 (CEACAM6/CD66c) Is a Protein Receptor for the Influenza a Virus. Viruses 2021; 13:v13050726. [PMID: 33919410 PMCID: PMC8143321 DOI: 10.3390/v13050726] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/10/2021] [Indexed: 12/24/2022] Open
Abstract
To establish a productive infection in host cells, viruses often use one or multiple host membrane glycoproteins as their receptors. For Influenza A virus (IAV) such a glycoprotein receptor has not been described, to date. Here we show that IAV is using the host membrane glycoprotein CD66c as a receptor for entry into human epithelial lung cells. Neuraminidase (NA), a viral spike protein, binds to CD66c on the cell surface during IAV entry into the host cells. Lung cells overexpressing CD66c showed an increase in virus binding and subsequent entry into the cell. Upon comparison, CD66c demonstrated higher binding capacity than other membrane glycoproteins (EGFR and DC-SIGN) reported earlier to facilitate IAV entry into host cells. siRNA mediated knockdown of CD66c from lung cells inhibited virus binding on cell surface and entry into cells. Blocking CD66c by antibody on the cell surface resulted in decreased virus entry. We found that CD66c is a specific glycoprotein receptor for influenza A virus that did not affect entry of non-IAV RNA virus (Hepatitis C virus). Finally, IAV pre-incubated with recombinant CD66c protein when administered intranasally in mice showed decreased cytopathic effects in mice lungs. This publication is the first to report CD66c (Carcinoembryonic cell adhesion molecule 6 or CEACAM6) as a glycoprotein receptor for Influenza A virus.
Collapse
|
24
|
Huang W, Li M, Luo G, Wu X, Su B, Zhao L, Zhang S, Chen X, Jia M, Zhu J, Su W, Zhang D. The Inflammatory Factors Associated with Disease Severity to Predict COVID-19 Progression. THE JOURNAL OF IMMUNOLOGY 2021; 206:1597-1608. [PMID: 33579725 DOI: 10.4049/jimmunol.2001327] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/26/2021] [Indexed: 01/08/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is associated with immune dysregulation and cytokine storm. Exploring the immune-inflammatory characteristics of COVID-19 patients is essential to reveal pathogenesis and predict progression. In this study, COVID-19 patients showed decreased CD3+, CD4+, and CD8+ T cells but increased neutrophils in circulation, exhibiting upregulated neutrophil-to-lymphocyte and neutrophil-to-CD8+ T cell ratio. IL-6, TNF-α, IL-1β, IL-18, IL-12/IL-23p40, IL-10, Tim-3, IL-8, neutrophil extracellular trap-related proteinase 3, and S100A8/A9 were elevated, whereas IFN-γ and C-type lectin domain family 9 member A (clec9A) were decreased in COVID-19 patients compared with healthy controls. When compared with influenza patients, the expressions of TNF-α, IL-18, IL-12/IL-23p40, IL-8, S100A8/A9 and Tim-3 were significantly increased in critical COVID-19 patients, and carcinoembryonic Ag, IL-8, and S100A8/A9 could serve as clinically available hematologic indexes for identifying COVID-19 from influenza. Moreover, IL-6, IL-8, IL-1β, TNF-α, proteinase 3, and S100A8/A9 were increased in bronchoalveolar lavage fluid of severe/critical patients compared with moderate patients, despite decreased CD4+ T cells, CD8+ T cells, B cells, and NK cells. Interestingly, bronchoalveolar IL-6, carcinoembryonic Ag, IL-8, S100A8/A9, and proteinase 3 were found to be predictive of COVID-19 severity and may serve as potential biomarkers for predicting COVID-19 progression and potential targets in therapeutic intervention of COVID-19.
Collapse
Affiliation(s)
- Wei Huang
- Department of Laboratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Mei Li
- Department of Laboratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Guangwei Luo
- Department of Respiratory and Critical Care Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Xiaojie Wu
- Department of Respiratory and Critical Care Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Bintao Su
- Department of Laboratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Lan Zhao
- Department of Laboratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Shuang Zhang
- Department of Laboratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Xiaofan Chen
- Department of Laboratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Min Jia
- Department of Laboratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Jianhua Zhu
- Department of Laboratory Medicine, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China
| | - Wen Su
- Department of Scientific Research, Wuhan No. 1 Hospital, Wuhan 430022, People's Republic of China; and
| | - Dongxin Zhang
- Department of Clinical Laboratory, Wuhan Fourth Hospital; Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430033, People's Republic of China
| |
Collapse
|
25
|
Acinetobacter baumannii Targets Human Carcinoembryonic Antigen-Related Cell Adhesion Molecules (CEACAMs) for Invasion of Pneumocytes. mSystems 2020; 5:5/6/e00604-20. [PMID: 33361319 PMCID: PMC7762790 DOI: 10.1128/msystems.00604-20] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multidrug-resistant Acinetobacter baumannii is regarded as a life-threatening pathogen mainly associated with nosocomial and community-acquired pneumonia. Here, we show that A. baumannii can bind the human carcinoembryonic antigen-related cell adhesion molecule (CEACAM) receptors CEACAM1, CEACAM5, and CEACAM6. This specific interaction enhances A. baumannii internalization in membrane-bound vacuoles, promptly decorated with Rab5, Rab7, and lipidated microtubule-associated protein light chain 3 (LC3). Dissecting intracellular signaling pathways revealed that infected pneumocytes trigger interleukin-8 (IL-8) secretion via the extracellular signal-regulated kinase (ERK)1/2 and nuclear factor-kappa B (NF-κB) signaling pathways for A. baumannii clearance. However, in CEACAM1-L-expressing cells, IL-8 secretion lasts only 24 h, possibly due to an A. baumannii-dependent effect on the CEACAM1-L intracellular domain. Conversely, the glycosylphosphatidylinositol-anchored CEACAM5 and CEACAM6 activate the c-Jun NH2-terminal kinase (JNK)1/2-Rubicon-NOX2 pathway, suggestive of LC3-associated phagocytosis. Overall, our data show for the first time novel mechanisms of adhesion to and invasion of pneumocytes by A. baumannii via CEACAM-dependent signaling pathways that eventually lead to bacterial killing. These findings suggest that CEACAM upregulation could put patients at increased risk of lower respiratory tract infection by A. baumannii IMPORTANCE This work shows for the first time that Acinetobacter baumannii binds to carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), CEACAM5, and CEACAM6. This binding significantly enhances A. baumannii internalization within alveolar host cell epithelia. Intracellular trafficking involves typical Rab5 and Rab7 vacuolar proteins as well as light chain 3 (LC3) and slowly progresses to bacterial killing by endosome acidification. CEACAM engagement by A. baumannii leads to distinct and specific downstream signaling pathways. The CEACAM1 pathway finely tunes interleukin-8 (IL-8) secretion, whereas CEACAM5 and CEACAM6 mediate LC3-associated phagocytosis. The present study provides new insights into A. baumannii-host interactions and could represent a promising therapeutic strategy to reduce pulmonary infections caused by this pathogen.
Collapse
|
26
|
DeLucia DC, Cardillo TM, Ang L, Labrecque MP, Zhang A, Hopkins JE, De Sarkar N, Coleman I, da Costa RMG, Corey E, True LD, Haffner MC, Schweizer MT, Morrissey C, Nelson PS, Lee JK. Regulation of CEACAM5 and Therapeutic Efficacy of an Anti-CEACAM5-SN38 Antibody-drug Conjugate in Neuroendocrine Prostate Cancer. Clin Cancer Res 2020; 27:759-774. [PMID: 33199493 DOI: 10.1158/1078-0432.ccr-20-3396] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/30/2020] [Accepted: 11/09/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Neuroendocrine prostate cancer (NEPC) is an aggressive form of castration-resistant prostate cancer (CRPC) for which effective therapies are lacking. We previously identified carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) as a promising NEPC cell surface antigen. Here we investigated the scope of CEACAM5 expression in end-stage prostate cancer, the basis for CEACAM5 enrichment in NEPC, and the therapeutic potential of the CEACAM5 antibody-drug conjugate labetuzumab govitecan in prostate cancer. EXPERIMENTAL DESIGN The expression of CEACAM5 and other clinically relevant antigens was characterized by multiplex immunofluorescence of a tissue microarray comprising metastatic tumors from 34 lethal metastatic CRPC (mCRPC) cases. A genetically defined neuroendocrine transdifferentiation assay of prostate cancer was developed to evaluate mechanisms of CEACAM5 regulation in NEPC. The specificity and efficacy of labetuzumab govitecan was determined in CEACAM5+ prostate cancer cell lines and patient-derived xenografts models. RESULTS CEACAM5 expression was enriched in NEPC compared with other mCRPC subtypes and minimally overlapped with prostate-specific membrane antigen, prostate stem cell antigen, and trophoblast cell surface antigen 2 expression. We focused on a correlation between the expression of the pioneer transcription factor ASCL1 and CEACAM5 to determine that ASCL1 can drive neuroendocrine reprogramming of prostate cancer which is associated with increased chromatin accessibility of the CEACAM5 core promoter and CEACAM5 expression. Labetuzumab govitecan induced DNA damage in CEACAM5+ prostate cancer cell lines and marked antitumor responses in CEACAM5+ CRPC xenograft models including chemotherapy-resistant NEPC. CONCLUSIONS Our findings provide insights into the scope and regulation of CEACAM5 expression in prostate cancer and strong support for clinical studies of labetuzumab govitecan for NEPC.
Collapse
Affiliation(s)
- Diana C DeLucia
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - Lisa Ang
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark P Labrecque
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Ailin Zhang
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - James E Hopkins
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Navonil De Sarkar
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ilsa Coleman
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Rui M Gil da Costa
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Eva Corey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Lawrence D True
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Michael C Haffner
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington
| | - Michael T Schweizer
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - Colm Morrissey
- Department of Urology, University of Washington School of Medicine, Seattle, Washington
| | - Peter S Nelson
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Urology, University of Washington School of Medicine, Seattle, Washington.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington.,Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| | - John K Lee
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington.,Department of Medicine, University of Washington School of Medicine, Seattle, Washington
| |
Collapse
|
27
|
Willcox MDP, Walsh K, Nichols JJ, Morgan PB, Jones LW. The ocular surface, coronaviruses and COVID-19. Clin Exp Optom 2020; 103:418-424. [PMID: 32406140 PMCID: PMC7272971 DOI: 10.1111/cxo.13088] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/24/2020] [Accepted: 04/22/2020] [Indexed: 12/21/2022] Open
Abstract
The ocular surface has been suggested as a site of infection with Coronavirus-2 (SARS-CoV-2) responsible for the coronavirus disease-19 (COVID-19). This review examines the evidence for this hypothesis, and its implications for clinical practice. Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), responsible for the COVID-19 pandemic, is transmitted by person-to-person contact, via airborne droplets, or through contact with contaminated surfaces. SARS-CoV-2 binds to angiotensin converting enzyme-2 (ACE2) to facilitate infection in humans. This review sets out to evaluate evidence for the ocular surface as a route of infection. A literature search in this area was conducted on 15 April 2020 using the Scopus database. In total, 287 results were returned and reviewed. There is preliminary evidence for ACE2 expression on corneal and conjunctival cells, but most of the other receptors to which coronaviruses bind appear to be found under epithelia of the ocular surface. Evidence from animal studies is limited, with a single study suggesting viral particles on the eye can travel to the lung, resulting in very mild infection. Coronavirus infection is rarely associated with conjunctivitis, with occasional cases reported in patients with confirmed COVID-19, along with isolated cases of conjunctivitis as a presenting sign. Coronaviruses have been rarely isolated from tears or conjunctival swabs. The evidence suggests coronaviruses are unlikely to bind to ocular surface cells to initiate infection. Additionally, hypotheses that the virus could travel from the nasopharynx or through the conjunctival capillaries to the ocular surface during infection are probably incorrect. Conjunctivitis and isolation of the virus from the ocular surface occur only rarely, and overwhelmingly in patients with confirmed COVID-19. Necessary precautions to prevent person-to-person transmission should be employed in clinical practice throughout the pandemic, and patients should be reminded to maintain good hygiene practices.
Collapse
Affiliation(s)
- Mark DP Willcox
- School of Optometry and Vision ScienceThe University of New South WalesSydneyNew South WalesAustralia
| | - Karen Walsh
- Centre for Ocular Research & Education (CORE), School of Optometry and Vision ScienceUniversity of WaterlooWaterlooOntarioCanada
| | - Jason J Nichols
- School of OptometryUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Philip B Morgan
- Eurolens Research, Division of Pharmacy and Optometry, The University of ManchesterManchesterUK
| | - Lyndon W Jones
- Centre for Ocular Research & Education (CORE), School of Optometry and Vision ScienceUniversity of WaterlooWaterlooOntarioCanada
| |
Collapse
|
28
|
Jeong S, Park MJ, Song W, Kim HS. Current immunoassay methods and their applications to clinically used biomarkers of breast cancer. Clin Biochem 2020; 78:43-57. [PMID: 32007438 DOI: 10.1016/j.clinbiochem.2020.01.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/13/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022]
Abstract
Breast cancer is the leading cause of cancer-related mortality worldwide, with a higher incidence in developed countries. The biomarkers for breast cancer such as estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2, CA (cancer antigen) 15-3, CA 27.29, and carcinoembryonic antigen have been recommended for use in the laboratory based on the guidelines of American and European societies. Immunoassays have been frequently and consistently used to detect these clinically established biomarkers of breast cancer. Despite the higher accessibility of serum biomarkers, including CA 15-3, CA 27.29, and CEA, compared to tissue markers, variations in immunoassays affect their standardization and clinical utility. When reviewing the immunoassays used to detect these serum markers, we found that the most frequently used immunoassay was enzyme-linked immunosorbent assay, followed by electrochemiluminescent immunoassay, and then chemiluminescence immunoassay for CA 15-3 and CEA. Meanwhile, the chemiluminescence immunoassay was the most common technique for CA27.29. The electrochemiluminescent immunoassay and monoclonal fluorometric assay have become the preferred methods in 2010-2019 compared to 2000-2009. Analytical and clinical performance factors such as sensitivity, specificity, detection limit, hazard risk to laboratory personnel, speed, and economic feasibility influenced these changes in user preference. When using the immunoassays, there should be a comprehensive understanding of the principles, advantages, vulnerability, and precautions for interpretation. In the future, a combination of immunological biomarkers and genetic platforms will benefit patients with breast cancer by facilitating prognosis prediction and guiding therapeutic intervention.
Collapse
Affiliation(s)
- Seri Jeong
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, 1 Singil-ro, Yeongdeungpo-gu, Seoul 07440, South Korea.
| | - Min-Jeong Park
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, 1 Singil-ro, Yeongdeungpo-gu, Seoul 07440, South Korea.
| | - Wonkeun Song
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, 1 Singil-ro, Yeongdeungpo-gu, Seoul 07440, South Korea.
| | - Hyon-Suk Kim
- Department of Laboratory Medicine, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, South Korea.
| |
Collapse
|
29
|
Klaile E, Müller MM, Zubiría-Barrera C, Brehme S, Klassert TE, Stock M, Durotin A, Nguyen TD, Feer S, Singer BB, Zipfel PF, Rudolphi S, Jacobsen ID, Slevogt H. Unaltered Fungal Burden and Lethality in Human CEACAM1-Transgenic Mice During Candida albicans Dissemination and Systemic Infection. Front Microbiol 2019; 10:2703. [PMID: 31849868 PMCID: PMC6889641 DOI: 10.3389/fmicb.2019.02703] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/07/2019] [Indexed: 12/29/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1, CD66a) is a receptor for Candida albicans. It is crucial for the immune response of intestinal epithelial cells to this opportunistic pathogen. Moreover, CEACAM1 is of importance for the mucosal colonization by different bacterial pathogens. We therefore studied the influence of the human CEACAM1 receptor in human CEACAM1-transgenic mice on the C. albicans colonization and infection utilizing a colonization/dissemination and a systemic infection mouse model. Our results showed no alterations in the host response between the transgenic mice and the wild-type littermates to the C. albicans infections. Both mouse strains showed comparable C. albicans colonization and mycobiota, similar fungal burdens in various organs, and a similar survival in the systemic infection model. Interestingly, some of the mice treated with anti-bacterial antibiotics (to prepare them for C. albicans colonization via oral infection) also showed a strong reduction in endogenous fungi instead of the normally observed increase in fungal numbers. This was independent of the expression of human CEACAM1. In the systemic infection model, the human CEACAM1 expression was differentially regulated in the kidneys and livers of Candida-infected transgenic mice. Notably, in the kidneys, a total loss of the largest human CEACAM1 isoform was observed. However, the overwhelming immune response induced in the systemic infection model likely covered any CEACAM1-specific effects in the transgenic animals. In vitro studies using bone marrow-derived neutrophils from both mouse strains also revealed no differences in their reaction to C. albicans. In conclusion, in contrast to bacterial pathogens interacting with CEACAM1 on different mucosal surfaces, the human CEACAM1-transgenic mice did not reveal a role of human CEACAM1 in the in vivo candidiasis models used here. Further studies and different approaches will be needed to reveal a putative role of CEACAM1 in the host response to C. albicans.
Collapse
Affiliation(s)
- Esther Klaile
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Mario M Müller
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Cristina Zubiría-Barrera
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Saskia Brehme
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Tilman E Klassert
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Magdalena Stock
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Adrian Durotin
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Tien D Nguyen
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Sabina Feer
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| | - Bernhard B Singer
- Medical Faculty, Institute of Anatomy, University Duisburg-Essen, Essen, Germany
| | - Peter F Zipfel
- Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany.,Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany
| | - Sven Rudolphi
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.,Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany.,Center for Sepsis Control and Care (CSCC), University Hospital Jena, Jena, Germany
| | - Ilse D Jacobsen
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.,Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute (HKI), Jena, Germany.,Center for Sepsis Control and Care (CSCC), University Hospital Jena, Jena, Germany
| | - Hortense Slevogt
- Host Septomics Group, Centre for Innovation Competence (ZIK) Septomics, University Hospital Jena, Jena, Germany
| |
Collapse
|
30
|
Djureinovic D, Pontén V, Landelius P, Al Sayegh S, Kappert K, Kamali-Moghaddam M, Micke P, Ståhle E. Multiplex plasma protein profiling identifies novel markers to discriminate patients with adenocarcinoma of the lung. BMC Cancer 2019; 19:741. [PMID: 31357969 PMCID: PMC6664554 DOI: 10.1186/s12885-019-5943-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 07/16/2019] [Indexed: 12/12/2022] Open
Abstract
Background The overall prognosis of non-small cell lung cancer (NSCLC) is poor, and currently only patients with localized disease are potentially curable. Therefore, preferably non-invasively determined biomarkers that detect NSCLC patients at early stages of the disease are of high clinical relevance. The aim of this study was to identify and validate novel protein markers in plasma using the highly sensitive DNA-assisted multiplex proximity extension assay (PEA) to discriminate NSCLC from other lung diseases. Methods Plasma samples were collected from a total of 343 patients who underwent surgical resection for different lung diseases, including 144 patients with lung adenocarcinoma (LAC), 68 patients with non-malignant lung disease, 83 patients with lung metastasis of colorectal cancers and 48 patients with typical carcinoid. One microliter of plasma was analyzed using PEA, allowing detection and quantification of 92 established cancer related proteins. The concentrations of the plasma proteins were compared between disease groups. Results The comparison between LAC and benign samples revealed significantly different plasma levels for four proteins; CXCL17, CEACAM5, VEGFR2 and ERBB3 (adjusted p-value < 0.05). A multi-parameter classifier was developed to discriminate between samples from LAC patients and from patients with non-malignant lung conditions. With a bootstrap aggregated decision tree algorithm (TreeBagger), a sensitivity of 93% and specificity of 64% was achieved to detect LAC in this risk population. Conclusions By applying the highly sensitive PEA, reliable protein profiles could be determined in microliter amounts of plasma. We further identified proteins that demonstrated different plasma concentration in defined disease groups and developed a signature that holds potential to be included in a screening assay for early lung cancer detection. Electronic supplementary material The online version of this article (10.1186/s12885-019-5943-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dijana Djureinovic
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden.
| | - Victor Pontén
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden
| | - Per Landelius
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Sahar Al Sayegh
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden
| | - Kai Kappert
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden
| | - Elisabeth Ståhle
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
31
|
New therapeutic targets for the prevention of infectious acute exacerbations of COPD: role of epithelial adhesion molecules and inflammatory pathways. Clin Sci (Lond) 2019; 133:1663-1703. [PMID: 31346069 DOI: 10.1042/cs20181009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/15/2022]
Abstract
Chronic respiratory diseases are among the leading causes of mortality worldwide, with the major contributor, chronic obstructive pulmonary disease (COPD) accounting for approximately 3 million deaths annually. Frequent acute exacerbations (AEs) of COPD (AECOPD) drive clinical and functional decline in COPD and are associated with accelerated loss of lung function, increased mortality, decreased health-related quality of life and significant economic costs. Infections with a small subgroup of pathogens precipitate the majority of AEs and consequently constitute a significant comorbidity in COPD. However, current pharmacological interventions are ineffective in preventing infectious exacerbations and their treatment is compromised by the rapid development of antibiotic resistance. Thus, alternative preventative therapies need to be considered. Pathogen adherence to the pulmonary epithelium through host receptors is the prerequisite step for invasion and subsequent infection of surrounding structures. Thus, disruption of bacterial-host cell interactions with receptor antagonists or modulation of the ensuing inflammatory profile present attractive avenues for therapeutic development. This review explores key mediators of pathogen-host interactions that may offer new therapeutic targets with the potential to prevent viral/bacterial-mediated AECOPD. There are several conceptual and methodological hurdles hampering the development of new therapies that require further research and resolution.
Collapse
|
32
|
The prognostic significance of preoperative tumor marker (CEA, CA15-3) elevation in breast cancer patients: data from the Korean Breast Cancer Society Registry. Breast Cancer Res Treat 2019; 177:669-678. [DOI: 10.1007/s10549-019-05357-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 07/09/2019] [Indexed: 12/19/2022]
|
33
|
Metzinger MN, Verghese C, Hamouda DM, Lenhard A, Choucair K, Senzer N, Brunicardi FC, Dworkin L, Nemunaitis J. Chimeric Antigen Receptor T-Cell Therapy: Reach to Solid Tumor Experience. Oncology 2019; 97:59-74. [PMID: 31261152 DOI: 10.1159/000500488] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 03/12/2019] [Indexed: 11/19/2022]
Abstract
Chimeric antigen receptor (CAR) modified T-cell therapy, a unique platform technology highlighting precision medicine through utilization of molecular biology and cell-based therapeutics has shown unprecedented rates in patients with hematological malignancies such as acute lymphocyte leukemia, non-Hodgkin's lymphoma and multiple myeloma (MM). With the approval of CD19-targeted CAR T-cells by the Food and Drug Administration in acute lymphoblastic leukemia (ALL) and NHL, this technology is positioned for aggressive expansion to combination therapeutic opportunities and proof of principle towards utility in other malignant disorders. However, despite the impressive results seen with hematological malignancies, CAR T-cells have shown limited efficacy in solid tumors with several unsuccessful preclinical studies. Regardless, these attempts have provided us with a better understanding of the imminent challenges specific to solid tumors even if they have not so far led to expanded clinical treatment opportunities outside ALL/NHL/MM. This review summarizes our current understanding of CAR T-cell mechanism of action, while presenting the major limitations of CAR T-cell derived treatments in solid tumors. We further discuss recent findings and present new potential strategies to overcome the challenges facing solid tumor targeting by CAR T-cell platforms.
Collapse
Affiliation(s)
- Matthew N Metzinger
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Cherian Verghese
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Danae M Hamouda
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Amanda Lenhard
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Khalil Choucair
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - Neil Senzer
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | | | - Lance Dworkin
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA
| | - John Nemunaitis
- University of Toledo College of Medicine and Life Sciences, Toledo, Ohio, USA, .,ProMedica Health System, Toledo, Ohio, USA,
| |
Collapse
|
34
|
Wang Z, Maschera B, Lea S, Kolsum U, Michalovich D, Van Horn S, Traini C, Brown JR, Hessel EM, Singh D. Airway host-microbiome interactions in chronic obstructive pulmonary disease. Respir Res 2019; 20:113. [PMID: 31170986 PMCID: PMC6555748 DOI: 10.1186/s12931-019-1085-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 05/28/2019] [Indexed: 12/31/2022] Open
Abstract
Background Little is known about the interactions between the lung microbiome and host response in chronic obstructive pulmonary disease (COPD). Methods We performed a longitudinal 16S ribosomal RNA gene-based microbiome survey on 101 sputum samples from 16 healthy subjects and 43 COPD patients, along with characterization of host sputum transcriptome and proteome in COPD patients. Results Dysbiosis of sputum microbiome was observed with significantly increased relative abundance of Moraxella in COPD versus healthy subjects and during COPD exacerbations, and Haemophilus in COPD ex-smokers versus current smokers. Multivariate modeling on sputum microbiome, host transcriptome and proteome profiles revealed that significant associations between Moraxella and Haemophilus, host interferon and pro-inflammatory signaling pathways and neutrophilic inflammation predominated among airway host-microbiome interactions in COPD. While neutrophilia was positively correlated with Haemophilus, interferon signaling was more strongly linked to Moraxella. Moreover, while Haemophilus was significantly associated with host factors both in stable state and during exacerbations, Moraxella-associated host responses were primarily related to exacerbations. Conclusions Our study highlights a significant airway host-microbiome interplay associated with COPD inflammation and exacerbations. These findings indicate that Haemophilus and Moraxella influence different components of host immune response in COPD, and that novel therapeutic strategies should consider targeting these bacteria and their associated host pathways in COPD. Electronic supplementary material The online version of this article (10.1186/s12931-019-1085-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhang Wang
- Computational Biology, Human Genetics, Research and Development (R&D), GlaxoSmithKline (GSK), 1250 S. Collegeville Road, Collegeville, PA, 19426-0989, USA.,Present Address: School of Life Sciences, South China Normal University, Guangzhou, 510631, People's Republic of China
| | - Barbara Maschera
- Refractory Respiratory Inflammation Discovery Performance Unit, Respiratory Therapy Area, R&D, GSK, Stevenage, SG1 2NY, UK
| | - Simon Lea
- University of Manchester and University Hospital of South Manchester, Manchester, M23 9QZ, UK
| | - Umme Kolsum
- University of Manchester and University Hospital of South Manchester, Manchester, M23 9QZ, UK
| | - David Michalovich
- Refractory Respiratory Inflammation Discovery Performance Unit, Respiratory Therapy Area, R&D, GSK, Stevenage, SG1 2NY, UK
| | - Stephanie Van Horn
- Functional Genomics, Medicinal Science and Technology, R&D, GSK, Collegeville, PA, 19426, USA
| | - Christopher Traini
- Functional Genomics, Medicinal Science and Technology, R&D, GSK, Collegeville, PA, 19426, USA
| | - James R Brown
- Computational Biology, Human Genetics, Research and Development (R&D), GlaxoSmithKline (GSK), 1250 S. Collegeville Road, Collegeville, PA, 19426-0989, USA.
| | - Edith M Hessel
- Refractory Respiratory Inflammation Discovery Performance Unit, Respiratory Therapy Area, R&D, GSK, Stevenage, SG1 2NY, UK
| | - Dave Singh
- University of Manchester and University Hospital of South Manchester, Manchester, M23 9QZ, UK
| |
Collapse
|
35
|
Bertuzzi M, Hayes GE, Bignell EM. Microbial uptake by the respiratory epithelium: outcomes for host and pathogen. FEMS Microbiol Rev 2019; 43:145-161. [PMID: 30657899 PMCID: PMC6435450 DOI: 10.1093/femsre/fuy045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
Intracellular occupancy of the respiratory epithelium is a useful pathogenic strategy facilitating microbial replication and evasion of professional phagocytes or circulating antimicrobial drugs. A less appreciated but growing body of evidence indicates that the airway epithelium also plays a crucial role in host defence against inhaled pathogens, by promoting ingestion and quelling of microorganisms, processes that become subverted to favour pathogen activities and promote respiratory disease. To achieve a deeper understanding of beneficial and deleterious activities of respiratory epithelia during antimicrobial defence, we have comprehensively surveyed all current knowledge on airway epithelial uptake of bacterial and fungal pathogens. We find that microbial uptake by airway epithelial cells (AECs) is a common feature of respiratory host-microbe interactions whose stepwise execution, and impacts upon the host, vary by pathogen. Amidst the diversity of underlying mechanisms and disease outcomes, we identify four key infection scenarios and use best-characterised host-pathogen interactions as prototypical examples of each. The emergent view is one in which effi-ciency of AEC-mediated pathogen clearance correlates directly with severity of disease outcome, therefore highlighting an important unmet need to broaden our understanding of the antimicrobial properties of respiratory epithelia and associated drivers of pathogen entry and intracellular fate.
Collapse
Affiliation(s)
- Margherita Bertuzzi
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
- Lydia Becker Institute of Immunology and Inflammation, Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre
| | - Gemma E Hayes
- Northern Devon Healthcare NHS Trust, North Devon District Hospital, Raleigh Park, Barnstaple EX31 4JB, UK
| | - Elaine M Bignell
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre, Core Technology Facility, Grafton Street, Manchester M13 9NT, UK
- Lydia Becker Institute of Immunology and Inflammation, Biology, Medicine and Health. The University of Manchester, Manchester Academic Health Science Centre
| |
Collapse
|
36
|
Fischer K, Doehn JM, Herr C, Lachner C, Heinrich A, Kershaw O, Voss M, Jacobson MH, Gruber AD, Clauss M, Witzenrath M, Bals R, Gutbier B, Slevogt H. Acute Moraxella catarrhalis Airway Infection of Chronically Smoke-Exposed Mice Increases Mechanisms of Emphysema Development: A Pilot Study. Eur J Microbiol Immunol (Bp) 2018; 8:128-134. [PMID: 30719329 PMCID: PMC6348706 DOI: 10.1556/1886.2018.00019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 09/07/2018] [Indexed: 12/20/2022] Open
Abstract
In chronic obstructive pulmonary disease (COPD), acute exacerbations and emphysema development are characteristics for disease pathology. COPD is complicated by infectious exacerbations with acute worsening of respiratory symptoms with Moraxella catarrhalis as one of the most frequent pathogens. Although cigarette smoke (CS) is the primary risk factor, additional molecular mechanisms for emphysema development induced by bacterial infections are incompletely understood. We investigated the impact of M. catarrhalis on emphysema development in CS exposed mice and asked whether an additional infection would induce a solubilization of pro-apoptotic and pro-inflammatory endothelial monocyte-activating-protein-2 (EMAPII) to exert its activities in the pulmonary microvas-culature and other parts of the lungs not exposed directly to CS. Mice were exposed to smoke (6 or 9 months) and/or infected with M. catarrhalis. Lungs, bronchoalveolar lavage fluid (BALF), and plasma were analyzed. CS exposure reduced ciliated area, caused rarefaction of the lungs, and induced apoptosis. EMAPII was increased independent of prior smoke exposure in BALF of infected mice. Importantly, acute M. catarrhalis infection increased release of matrixmetalloproteases-9 and -12, which are involved in emphysema development and comprise a mechanism of EMAPII release. Our data suggest that acute M. catarrhalis infection represents an independent risk factor for emphysema development in smoke-exposed mice.
Collapse
Affiliation(s)
- Katja Fischer
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany.,Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Jan-Moritz Doehn
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany
| | - Christian Herr
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, University of the Saarland, Homburg Saar, Germany
| | - Carolin Lachner
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Annina Heinrich
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Olivia Kershaw
- Department of Veterinary Pathology, Freie Universität, Berlin, Germany
| | - Meike Voss
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, University of the Saarland, Homburg Saar, Germany
| | - Max H Jacobson
- Pathology and Laboratory Medicine, IU School of Medicine, Indianapolis, Indiana, USA
| | - Achim D Gruber
- Department of Veterinary Pathology, Freie Universität, Berlin, Germany
| | - Matthias Clauss
- Indiana Center for Vascular Biology and Medicine and Department of Cellular and Integrative Physiology, Indiana University, Indianapolis, Indiana, USA
| | - Martin Witzenrath
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Division of Pulmonary Inflammation, Berlin, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology, Respiratory Intensive Care Medicine, University of the Saarland, Homburg Saar, Germany
| | - Birgitt Gutbier
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Infectious Diseases and Respiratory Medicine, Berlin, Germany.,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Division of Pulmonary Inflammation, Berlin, Germany
| | - Hortense Slevogt
- Septomics Research Center, Jena University Hospital, Jena, Germany
| |
Collapse
|
37
|
Ye S, Cowled CJ, Yap CH, Stambas J. Deep sequencing of primary human lung epithelial cells challenged with H5N1 influenza virus reveals a proviral role for CEACAM1. Sci Rep 2018; 8:15468. [PMID: 30341336 PMCID: PMC6195505 DOI: 10.1038/s41598-018-33605-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/25/2018] [Indexed: 12/26/2022] Open
Abstract
Current prophylactic and therapeutic strategies targeting human influenza viruses include vaccines and antivirals. Given variable rates of vaccine efficacy and antiviral resistance, alternative strategies are urgently required to improve disease outcomes. Here we describe the use of HiSeq deep sequencing to analyze host gene expression in primary human alveolar epithelial type II cells infected with highly pathogenic avian influenza H5N1 virus. At 24 hours post-infection, 623 host genes were significantly upregulated, including the cell adhesion molecule CEACAM1. H5N1 virus infection stimulated significantly higher CEACAM1 protein expression when compared to influenza A PR8 (H1N1) virus, suggesting a key role for CEACAM1 in influenza virus pathogenicity. Furthermore, silencing of endogenous CEACAM1 resulted in reduced levels of proinflammatory cytokine/chemokine production, as well as reduced levels of virus replication following H5N1 infection. Our study provides evidence for the involvement of CEACAM1 in a clinically relevant model of H5N1 infection and may assist in the development of host-oriented antiviral strategies.
Collapse
Affiliation(s)
- Siying Ye
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia. .,AAHL CSIRO Deakin Collaborative Biosecurity Laboratory, East Geelong, Victoria, Australia.
| | | | - Cheng-Hon Yap
- University Hospital Geelong, Barwon Health, Geelong, Victoria, Australia
| | - John Stambas
- School of Medicine, Deakin University, Waurn Ponds, Victoria, Australia.,AAHL CSIRO Deakin Collaborative Biosecurity Laboratory, East Geelong, Victoria, Australia
| |
Collapse
|
38
|
Suda T, Tatsumi T, Nishio A, Kegasawa T, Yoshioka T, Yamada R, Furuta K, Kodama T, Shigekawa M, Hikita H, Sakamori R, Fukuhara T, Matsuura Y, Takehara T. CEACAM1 Is Associated With the Suppression of Natural Killer Cell Function in Patients With Chronic Hepatitis C. Hepatol Commun 2018; 2:1247-1258. [PMID: 30288478 PMCID: PMC6167072 DOI: 10.1002/hep4.1240] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 05/20/2018] [Indexed: 12/15/2022] Open
Abstract
Natural killer cells (NK cells) play an essential role in the immunological mechanism underlying chronic hepatitis C (CHC). Impairment of NK cell function facilitates persistent infection with hepatitis C virus (HCV) and hepatocellular carcinogenesis. However, the mechanism by which NK cell activity is suppressed in CHC is not completely understood. In this study, we focused on carcinoembryonic antigen–related cell‐adhesion molecule 1 (CEACAM1). CEACAM1 is thought to suppress NK cell function. We examined the effect of CEACAM1 on NK cell function in CHC. We investigated the function of CEACAM1 in vitro using Huh7.5.1 cells and the HCV‐Japanese fulminant hepatitis (JFH)‐1 strain. We analyzed serum CEACAM1 level, NK cell function, and CEACAM1 messenger RNA (mRNA) level in human liver samples. Levels of CEACAM1 on the cell surface, CEACAM1 mRNA levels, and soluble CEACAM1 levels in supernatants were significantly higher in Huh7.5.1 cells infected with JFH‐1 (Huh7.5.1/JFH‐1 cells) than in Huh7.5.1 cells. Significantly higher NK cell cytotoxicity was observed toward K562 cells after coculture with CEACAM1 knockout Huh7.5.1/JFH‐1 cells than after coculture with Huh7.5.1/JFH‐1 cells. CEACAM1 expression was induced by the HCV E2 glycoprotein in HCV infection. Significantly higher serum CEACAM1 levels were detected in patients with CHC compared with healthy subjects and patients who achieved sustained virological responses. The expression of CD107a on NK cells from patients with CHC was negatively correlated with serum CEACAM1 levels. Significantly higher levels of CEACAM1 mRNA were detected in HCV‐infected livers compared with uninfected livers. Conclusion: CEACAM1 expression was induced in hepatocytes following HCV infection and decreased NK cell cytotoxicity. These results demonstrate a possible role for CEACAM1 in the pathogenesis of CHC and hepatocellular carcinoma progression.
Collapse
Affiliation(s)
- Takahiro Suda
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Tomohide Tatsumi
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Akira Nishio
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Tadashi Kegasawa
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Teppei Yoshioka
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Ryoko Yamada
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Kunimaro Furuta
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Takahiro Kodama
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Minoru Shigekawa
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Ryotaro Sakamori
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| | - Takasuke Fukuhara
- Department of Molecular Virology Research Institute for Microbial Diseases, Osaka University Suita Japan
| | - Yoshiharu Matsuura
- Department of Molecular Virology Research Institute for Microbial Diseases, Osaka University Suita Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology Osaka University Graduate School of Medicine Suita Japan
| |
Collapse
|
39
|
Bednarek K, Kostrzewska-Poczekaj M, Szaumkessel M, Kiwerska K, Paczkowska J, Byzia E, Ustaszewski A, Janiszewska J, Bartochowska A, Grenman R, Wierzbicka M, Szyfter K, Giefing M, Jarmuz-Szymczak M. Downregulation of CEACAM6 gene expression in laryngeal squamous cell carcinoma is an effect of DNA hypermethylation and correlates with disease progression. Am J Cancer Res 2018; 8:1249-1261. [PMID: 30094098 PMCID: PMC6079150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 05/27/2018] [Indexed: 06/08/2023] Open
Abstract
We have turned our attention to CEACAM6 gene, already described as deregulated in various types of cancer. By using the expression microarrays performed on the set of 16 laryngeal squamous cell carcinoma (LSCC) samples: 11 cell lines and 5 primary tumors we have shown downregulation of CEACAM6 gene as compared to non cancer controls from head and neck region. CEACAM6 gene downregulation, further confirmed by quantitative PCR on 25 LSCC cell lines, was observed in cell lines derived from recurrent tumors in comparison to controls. A significant gene downregulation was observed in cell lines derived from advanced, high grade tumors in comparison to controls. Intrigued by the recurrent transcriptional loss of CEACAM6 we searched for the mechanism potentially responsible for its downregulation and hence we analyzed DNA copy number changes (a-CGH), promoter DNA methylation status and occurrence of gene mutations (in silico). Neither the analysis of gene copy number, nor the mutation screen has shown recurrent deletions or mutations, that could contribute to the observed downregulation of the gene. However, by using bisulfite pyrosequencing, we have shown DNA hypermethylation (mean DNA methylation > 78%) of CEACAM6 promoter region in 9/25 (36%) LSCC cell lines. Importantly, the 5-aza-2-deoxycytidine-induced inhibition of DNA methylation resulted in restoration of CEACAM6 expression in the two LSCC cell lines on mRNA level. In summary, we have shown that recurrent downregulation of CEACAM6 in LSCC is dependent on the gene's promoter DNA methylation and is observed predominantly in large, poorly differentiated tumors and recurrences.
Collapse
Affiliation(s)
- Kinga Bednarek
- Institute of Human Genetics, Polish Academy of SciencesPoznan, Poland
| | | | | | - Katarzyna Kiwerska
- Institute of Human Genetics, Polish Academy of SciencesPoznan, Poland
- Department of Tumor Pathology, Greater Poland Cancer CenterPoznan, Poland
| | - Julia Paczkowska
- Institute of Human Genetics, Polish Academy of SciencesPoznan, Poland
| | - Ewa Byzia
- Institute of Human Genetics, Polish Academy of SciencesPoznan, Poland
| | - Adam Ustaszewski
- Institute of Human Genetics, Polish Academy of SciencesPoznan, Poland
| | | | - Anna Bartochowska
- Department of Otolaryngology and Laryngological Oncology, Poznan University of Medical SciencesPoznan, Poland
| | - Reidar Grenman
- Department of Otorhinolaryngology-Head and Neck Surgery and Department of Medical Biochemistry, Turku University Hospital and University of TurkuTurku, Finland
| | - Malgorzata Wierzbicka
- Institute of Human Genetics, Polish Academy of SciencesPoznan, Poland
- Department of Otolaryngology and Laryngological Oncology, Poznan University of Medical SciencesPoznan, Poland
| | - Krzysztof Szyfter
- Institute of Human Genetics, Polish Academy of SciencesPoznan, Poland
| | - Maciej Giefing
- Institute of Human Genetics, Polish Academy of SciencesPoznan, Poland
- Department of Otolaryngology and Laryngological Oncology, Poznan University of Medical SciencesPoznan, Poland
| | - Malgorzata Jarmuz-Szymczak
- Institute of Human Genetics, Polish Academy of SciencesPoznan, Poland
- Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical SciencesPoznan, Poland
| |
Collapse
|
40
|
Moonens K, Hamway Y, Neddermann M, Reschke M, Tegtmeyer N, Kruse T, Kammerer R, Mejías-Luque R, Singer BB, Backert S, Gerhard M, Remaut H. Helicobacter pylori adhesin HopQ disrupts trans dimerization in human CEACAMs. EMBO J 2018; 37:embj.201798665. [PMID: 29858229 DOI: 10.15252/embj.201798665] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/08/2018] [Accepted: 05/15/2018] [Indexed: 01/24/2023] Open
Abstract
The human gastric pathogen Helicobacter pylori is a major causative agent of gastritis, peptic ulcer disease, and gastric cancer. As part of its adhesive lifestyle, the bacterium targets members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family by the conserved outer membrane adhesin HopQ. The HopQ-CEACAM1 interaction is associated with inflammatory responses and enables the intracellular delivery and phosphorylation of the CagA oncoprotein via a yet unknown mechanism. Here, we generated crystal structures of HopQ isotypes I and II bound to the N-terminal domain of human CEACAM1 (C1ND) and elucidated the structural basis of H. pylori specificity toward human CEACAM receptors. Both HopQ alleles target the β-strands G, F, and C of C1ND, which form the trans dimerization interface in homo- and heterophilic CEACAM interactions. Using SAXS, we show that the HopQ ectodomain is sufficient to induce C1ND monomerization and thus providing H. pylori a route to influence CEACAM-mediated cell adherence and signaling events.
Collapse
Affiliation(s)
- Kristof Moonens
- Structural and Molecular Microbiology, Structural Biology Research Center, VIB, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Youssef Hamway
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Matthias Neddermann
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen, Erlangen, Germany
| | - Marc Reschke
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Nicole Tegtmeyer
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen, Erlangen, Germany
| | | | - Robert Kammerer
- Institute of Immunology, Friedrich-Loeffler Institut, Greifswald-Insel Riems, Germany
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Steffen Backert
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen, Erlangen, Germany
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Han Remaut
- Structural and Molecular Microbiology, Structural Biology Research Center, VIB, Brussels, Belgium .,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
41
|
Lehmann R, Müller MM, Klassert TE, Driesch D, Stock M, Heinrich A, Conrad T, Moore C, Schier UK, Guthke R, Slevogt H. Differential regulation of the transcriptomic and secretomic landscape of sensor and effector functions of human airway epithelial cells. Mucosal Immunol 2018; 11:627-642. [PMID: 29297499 DOI: 10.1038/mi.2017.100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 10/17/2017] [Indexed: 02/04/2023]
Abstract
Protein secretion upon TLR, TNFR1, and IFNGR ligation in the human airways is considered to be central for the orchestration of pulmonary inflammatory and immune responses. In this study, we compared the gene expression and protein secretion profiles in response to specific stimulation of all expressed TLRs and in further comparison to TNFR1 and IFNGR in primary human airway epithelial cells. In addition to 22 cytokines, we observed the receptor-induced regulation of 571 genes and 1,012 secreted proteins. Further analysis revealed high similarities between the transcriptional TLR sensor and TNFR1 effector responses. However, secretome to transcriptome comparisons showed a broad receptor stimulation-dependent release of proteins that were not transcriptionally regulated. Many of these proteins are annotated to exosomes with associations to, for example, antigen presentation and wound-healing, or were identified as secretable proteins related to immune responses. Thus, we show a hitherto unrecognized scope of receptor-induced responses in airway epithelium, involving several additional functions for the immune response, exosomal communication and tissue homeostasis.
Collapse
Affiliation(s)
- Roland Lehmann
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Mario M Müller
- Septomics Research Centre, Jena University Hospital, Jena, Germany
- Integrated Research and Treatment Centre - Centre for Sepsis Control and Care (CSCC), Jena University Hospital, Jena, Germany
| | | | | | - Magdalena Stock
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Anina Heinrich
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Theresia Conrad
- Septomics Research Centre, Jena University Hospital, Jena, Germany
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| | - Christoph Moore
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Uta K Schier
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| | - Reinhard Guthke
- Research Group Systems Biology and Bioinformatics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute, Jena, Germany
| | - Hortense Slevogt
- Septomics Research Centre, Jena University Hospital, Jena, Germany
| |
Collapse
|
42
|
Singhania A, Wallington JC, Smith CG, Horowitz D, Staples KJ, Howarth PH, Gadola SD, Djukanović R, Woelk CH, Hinks TSC. Multitissue Transcriptomics Delineates the Diversity of Airway T Cell Functions in Asthma. Am J Respir Cell Mol Biol 2018; 58:261-270. [PMID: 28933920 DOI: 10.1165/rcmb.2017-0162oc] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Asthma arises from the complex interplay of inflammatory pathways in diverse cell types and tissues. We sought to undertake a comprehensive transcriptomic assessment of the epithelium and airway T cells that remain understudied in asthma and investigate interactions between multiple cells and tissues. Epithelial brushings and flow-sorted CD3+ T cells from sputum and BAL were obtained from healthy subjects (n = 19) and patients with asthma (mild, moderate, and severe asthma; n = 46). Gene expression was assessed using Affymetrix HT HG-U133+ PM GeneChips, and results were validated by real-time quantitative PCR. In the epithelium, IL-13 response genes (POSTN, SERPINB2, and CLCA1), mast cell mediators (CPA3 and TPSAB1), inducible nitric oxide synthase, and cystatins (CST1, CST2, and CST4) were upregulated in mild asthma, but, except for cystatins, were suppressed by corticosteroids in moderate asthma. In severe asthma-with predominantly neutrophilic phenotype-several distinct processes were upregulated, including neutrophilia (TCN1 and MMP9), mucins, and oxidative stress responses. The majority of the disease signature was evident in sputum T cells in severe asthma, where 267 genes were differentially regulated compared with health, highlighting compartmentalization of inflammation. This signature included IL-17-inducible chemokines (CXCL1, CXCL2, CXCL3, IL8, and CSF3) and chemoattractants for neutrophils (IL8, CCL3, and LGALS3), T cells, and monocytes. A protein interaction network in severe asthma highlighted signatures of responses to bacterial infections across tissues (CEACAM5, CD14, and TLR2), including Toll-like receptor signaling. In conclusion, the activation of innate immune pathways in the airways suggests that activated T cells may be driving neutrophilic inflammation and steroid-insensitive IL-17 response in severe asthma.
Collapse
Affiliation(s)
- Akul Singhania
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and
| | - Joshua C Wallington
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and
| | - Caroline G Smith
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,2 National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Daniel Horowitz
- 3 Janssen Research and Development, High Wycombe, Buckinghamshire, United Kingdom
| | - Karl J Staples
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and
| | - Peter H Howarth
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,2 National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Stephan D Gadola
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,4 Roche, F. Hoffman-La Roche AG, Konzern-Hauptsitz, Basel, Switzerland; and
| | - Ratko Djukanović
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,2 National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom
| | - Christopher H Woelk
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and
| | - Timothy S C Hinks
- 1 Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, and.,2 National Institute for Health Research Southampton Respiratory Biomedical Research Unit, Southampton University Hospital, Southampton, United Kingdom.,5 Nuffield Department of Medicine and National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, United Kingdom
| |
Collapse
|
43
|
Abstract
CEA TCB is a novel T-cell-bispecific (TCB) antibody targeting the carcinoembryonic antigen (CEA) expressed on tumor cells and the CD3 epsilon chain (CD3e) present on T cells, which is currently in Phase 1 clinical trials (NCT02324257) for the treatment of CEA-positive solid tumors. Because the human CEA (hCEA) binder of CEA TCB does not cross-react with cynomolgus monkey and CEA is absent in rodents, alternative nonclinical safety evaluation approaches were considered. These included the development of a cynomolgus monkey cross-reactive homologous (surrogate) antibody (cyCEA TCB) for its evaluation in cynomolgus monkey and the development of double-transgenic mice, expressing hCEA and human CD3e (hCEA/hCD3e Tg), as a potential alternative species for nonclinical safety studies. However, a battery of nonclinical in vitro/ex vivo experiments demonstrated that neither of the previous approaches provided a suitable and pharmacologically relevant model to assess the safety of CEA TCB. Therefore, an alternative approach, a minimum anticipated biological effect level (MABEL), based on an in vitro tumor lysis assay was used to determine the starting dose for the first-in-human study. Using the most conservative approach to the MABEL assessment, a dose of 52 μg was selected as a safe starting dose for clinical study.
Collapse
|
44
|
The clinical efficacy of first-generation carcinoembryonic antigen (CEACAM5)-specific CAR T cells is limited by poor persistence and transient pre-conditioning-dependent respiratory toxicity. Cancer Immunol Immunother 2017; 66:1425-1436. [PMID: 28660319 PMCID: PMC5645435 DOI: 10.1007/s00262-017-2034-7] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/18/2017] [Indexed: 12/12/2022]
Abstract
The primary aim of this clinical trial was to determine the feasibility of delivering first-generation CAR T cell therapy to patients with advanced, CEACAM5+ malignancy. Secondary aims were to assess clinical efficacy, immune effector function and optimal dose of CAR T cells. Three cohorts of patients received increasing doses of CEACAM5+-specific CAR T cells after fludarabine pre-conditioning plus systemic IL2 support post T cell infusion. Patients in cohort 4 received increased intensity pre-conditioning (cyclophosphamide and fludarabine), systemic IL2 support and CAR T cells. No objective clinical responses were observed. CAR T cell engraftment in patients within cohort 4 was significantly higher. However, engraftment was short-lived with a rapid decline of systemic CAR T cells within 14 days. Patients in cohort 4 had transient, acute respiratory toxicity which, in combination with lack of prolonged CAR T cell persistence, resulted in the premature closure of the trial. Elevated levels of systemic IFNγ and IL-6 implied that the CEACAM5-specific T cells had undergone immune activation in vivo but only in patients receiving high-intensity pre-conditioning. Expression of CEACAM5 on lung epithelium may have resulted in this transient toxicity. Raised levels of serum cytokines including IL-6 in these patients implicate cytokine release as one of several potential factors exacerbating the observed respiratory toxicity. Whilst improved CAR designs and T cell production methods could improve the systemic persistence and activity, methods to control CAR T ‘on-target, off-tissue’ toxicity are required to enable a clinical impact of this approach in solid malignancies.
Collapse
|
45
|
Schauer AE, Klassert TE, von Lachner C, Riebold D, Schneeweiß A, Stock M, Müller MM, Hammerschmidt S, Bufler P, Seifert U, Dietert K, Dinarello CA, Nold MF, Gruber AD, Nold-Petry CA, Slevogt H. IL-37 Causes Excessive Inflammation and Tissue Damage in Murine Pneumococcal Pneumonia. J Innate Immun 2017; 9:403-418. [PMID: 28601872 PMCID: PMC6738772 DOI: 10.1159/000469661] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/09/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
Streptococcus pneumoniae infections can lead to severe complications with excessive immune activation and tissue damage. Interleukin-37 (IL-37) has gained importance as a suppressor of innate and acquired immunity, and its effects have been therapeutic as they prevent tissue damage in autoimmune and inflammatory diseases. By using RAW macrophages, stably transfected with human IL-37, we showed a 70% decrease in the cytokine levels of IL-6, TNF-α, and IL-1β, and a 2.2-fold reduction of the intracellular killing capacity of internalized pneumococci in response to pneumococcal infection. In a murine model of infection with S. pneumoniae, using mice transgenic for human IL-37b (IL-37tg), we observed an initial decrease in cytokine expression of IL-6, TNF-α, and IL-1β in the lungs, followed by a late-phase enhancement of pneumococcal burden and subsequent increase of proinflammatory cytokine levels. Additionally, a marked increase in recruitment of alveolar macrophages and neutrophils was noted, while TRAIL mRNA was reduced 3-fold in lungs of IL-37tg mice, resulting in necrotizing pneumonia with augmented death of infiltrating neutrophils, enhanced bacteremic spread, and increased mortality. In conclusion, we have identified that IL-37 modulates several core components of a successful inflammatory response to pneumococcal pneumonia, which lead to increased inflammation, tissue damage, and mortality.
Collapse
Affiliation(s)
- Anja E. Schauer
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | | | | | - Diana Riebold
- InfectoGnostics Research Campus Jena, Centre for Applied Research Jena, Jena, Germany
| | - Anne Schneeweiß
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Magdalena Stock
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Mario M. Müller
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Sven Hammerschmidt
- Department of Genetics of Microorganisms, Interfaculty Institute for Genetics and Functional Genomics, Ernst Moritz Arndt University of Greifswald, Greifswald, Germany
| | - Philip Bufler
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Ulrike Seifert
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Kristina Dietert
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, USA
- Department of Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel F. Nold
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Achim D. Gruber
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Claudia A. Nold-Petry
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia
| | - Hortense Slevogt
- Septomics Research Center, Jena University Hospital, Jena, Germany
| |
Collapse
|
46
|
Kc R, Shukla SD, Walters EH, O'Toole RF. Temporal upregulation of host surface receptors provides a window of opportunity for bacterial adhesion and disease. MICROBIOLOGY-SGM 2017; 163:421-430. [PMID: 28113047 DOI: 10.1099/mic.0.000434] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Host surface receptors provide bacteria with a foothold from which to attach, colonize and, in some cases, invade tissue and elicit human disease. In this review, we discuss several key host receptors and cognate adhesins that function in bacterial pathogenesis. In particular, we examine the elevated expression of host surface receptors such as CEACAM-1, CEACAM-6, ICAM-1 and PAFR in response to specific stimuli. We explore how upregulated receptors, in turn, expose the host to a range of bacterial infections in the respiratory tract. It is apparent that exploitation of receptor induction for bacterial adherence is not unique to one body system, but is also observed in the central nervous, gastrointestinal and urogenital systems. Prokaryotic pathogens which utilize this mechanism for their infectivity include Streptococcus pneumoniae, Haemophilus influenzae, Neisseria meningitidis and Escherichia coli. A number of approaches have been used, in both in vitro and in vivo experimental models, to inhibit bacterial attachment to temporally expressed host receptors. Some of these novel strategies may advance future targeted interventions for the prevention and treatment of bacterial disease.
Collapse
Affiliation(s)
- Rajendra Kc
- School of Medicine, Faculty of Health, University of Tasmania, Hobart, TAS 7000, Australia
| | - Shakti D Shukla
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Newcastle, NSW 2308, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW 2305, Australia
| | - Eugene H Walters
- School of Medicine, Faculty of Health, University of Tasmania, Hobart, TAS 7000, Australia
| | - Ronan F O'Toole
- School of Medicine, Faculty of Health, University of Tasmania, Hobart, TAS 7000, Australia
| |
Collapse
|
47
|
|
48
|
|
49
|
Binding of Candida albicans to Human CEACAM1 and CEACAM6 Modulates the Inflammatory Response of Intestinal Epithelial Cells. mBio 2017; 8:mBio.02142-16. [PMID: 28292985 PMCID: PMC5350469 DOI: 10.1128/mbio.02142-16] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Candida albicans colonizes human mucosa, including the gastrointestinal tract, as a commensal. In immunocompromised patients, C. albicans can breach the intestinal epithelial barrier and cause fatal invasive infections. Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1; CD66a), CEACAM5 (CEA), and CEACAM6 (CD66c) are immunomodulatory receptors expressed on human mucosa and are recruited by bacterial and viral pathogens. Here we show for the first time that a fungal pathogen (i.e., C. albicans) also binds directly to the extracellular domain of human CEACAM1, CEACAM3, CEACAM5, and CEACAM6. Binding was specific for human CEACAMs and mediated by the N-terminal IgV-like domain. In enterocytic C2BBe1 cells, C. albicans caused a transient tyrosine phosphorylation of CEACAM1 and induced higher expression of membrane-bound CEACAM1 and soluble CEACAM6. Lack of the CEACAM1 receptor after short hairpin RNA (shRNA) knockdown abolished CXCL8 (interleukin-8) secretion by C2BBe1 cells in response to C. albicans In CEACAM1-competent cells, the addition of recombinant soluble CEACAM6 reduced the C. albicans-induced CXCL8 secretion.IMPORTANCE The present study demonstrates for the first time that fungal pathogens can be recognized by at least four members of the immunomodulatory CEACAM receptor family: CEACAM1, -3, -5, and -6. Three of the four receptors (i.e., CEACAM1, -5, and -6) are expressed in mucosal cells of the intestinal tract, where they are implicated in immunomodulation and control of tissue homeostasis. Importantly, the interaction of the major fungal pathogen in humans Candida albicans with CEACAM1 and CEACAM6 resulted in an altered epithelial immune response. With respect to the broad impact of CEACAM receptors on various aspects of the innate and the adaptive immune responses, in particular epithelial, neutrophil, and T cell behavior, understanding the role of CEACAMs in the host response to fungal pathogens might help to improve management of superficial and systemic fungal infections.
Collapse
|
50
|
Shikotra A, Choy DF, Siddiqui S, Arthur G, Nagarkar DR, Jia G, Wright AKA, Ohri CM, Doran E, Butler CA, Hargadon B, Abbas AR, Jackman J, Wu LC, Heaney LG, Arron JR, Bradding P. A CEACAM6-High Airway Neutrophil Phenotype and CEACAM6-High Epithelial Cells Are Features of Severe Asthma. THE JOURNAL OF IMMUNOLOGY 2017; 198:3307-3317. [PMID: 28275137 DOI: 10.4049/jimmunol.1600606] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 02/06/2017] [Indexed: 12/27/2022]
Abstract
Severe asthma represents a major unmet clinical need; understanding the pathophysiology is essential for the development of new therapies. Using microarray analysis, we previously found three immunological clusters in asthma: Th2-high, Th17-high, and Th2/17-low. Although new therapies are emerging for Th2-high disease, identifying molecular pathways in Th2-low disease remains an important goal. Further interrogation of our previously described microarray dataset revealed upregulation of gene expression for carcinoembryonic Ag cell adhesion molecule (CEACAM) family members in the bronchi of patients with severe asthma. Our aim was therefore to explore the distribution and cellular localization of CEACAM6 using immunohistochemistry on bronchial biopsy tissue obtained from patients with mild-to-severe asthma and healthy control subjects. Human bronchial epithelial cells were used to investigate cytokine and corticosteroid in vitro regulation of CEACAM6 gene expression. CEACAM6 protein expression in bronchial biopsies was increased in airway epithelial cells and lamina propria inflammatory cells in severe asthma compared with healthy control subjects. CEACAM6 in the lamina propria was localized to neutrophils predominantly. Neutrophil density in the bronchial mucosa was similar across health and the spectrum of asthma severity, but the percentage of neutrophils expressing CEACAM6 was significantly increased in severe asthma, suggesting the presence of an altered neutrophil phenotype. CEACAM6 gene expression in cultured epithelial cells was upregulated by wounding and neutrophil elastase. In summary, CEACAM6 expression is increased in severe asthma and primarily associated with airway epithelial cells and tissue neutrophils. CEACAM6 may contribute to the pathology of treatment-resistant asthma via neutrophil and airway epithelial cell-dependent pathways.
Collapse
Affiliation(s)
- Aarti Shikotra
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester LE3 9QP, United Kingdom
| | | | - Salman Siddiqui
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester LE3 9QP, United Kingdom
| | - Greer Arthur
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester LE3 9QP, United Kingdom
| | | | - Guiquan Jia
- Genentech, Inc., South San Francisco, CA 94080
| | - Adam K A Wright
- University Hospitals of Leicester National Health Service Trust, Glenfield Hospital, Leicester LE3 9QP, United Kingdom; and
| | - Chandra M Ohri
- University Hospitals of Leicester National Health Service Trust, Glenfield Hospital, Leicester LE3 9QP, United Kingdom; and
| | - Emma Doran
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, United Kingdom
| | - Claire A Butler
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, United Kingdom
| | - Beverley Hargadon
- University Hospitals of Leicester National Health Service Trust, Glenfield Hospital, Leicester LE3 9QP, United Kingdom; and
| | | | | | - Lawren C Wu
- Genentech, Inc., South San Francisco, CA 94080
| | - Liam G Heaney
- Centre for Infection and Immunity, Queen's University Belfast, Belfast BT9 7AE, United Kingdom
| | | | - Peter Bradding
- Department of Infection, Immunity and Inflammation, Institute for Lung Health, University of Leicester, Leicester LE3 9QP, United Kingdom;
| |
Collapse
|