1
|
Liu Z, Zhao C, Yu H, Zhang R, Xue X, Jiang Z, Ge Z, Xu Y, Zhang W, Lin L, Chen Z. MCP-3 as a prognostic biomarker for severe fever with thrombocytopenia syndrome: a longitudinal cytokine profile study. Front Immunol 2024; 15:1379114. [PMID: 38812521 PMCID: PMC11134196 DOI: 10.3389/fimmu.2024.1379114] [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: 01/30/2024] [Accepted: 04/22/2024] [Indexed: 05/31/2024] Open
Abstract
Introduction Severe fever with thrombocytopenia syndrome (SFTS) is characterized by a high mortality rate and is associated with immune dysregulation. Cytokine storms may play an important role in adverse disease regression, this study aimed to assess the validity of MCP-3 in predicting adverse outcomes in SFTS patients and to investigate the longitudinal cytokine profile in SFTS patients. Methods The prospective study was conducted at Yantai Qishan Hospital from May to November 2022. We collected clinical data and serial blood samples during hospitalization, patients with SFTS were divided into survival and non-survival groups based on the clinical prognosis. Results The levels of serum 48 cytokines were measured using Luminex assays. Compared to healthy controls, SFTS patients exhibited higher levels of most cytokines. The non-survival group had significantly higher levels of 32 cytokines compared to the survival group. Among these cytokines, MCP-3 was ranked as the most significant variable by the random forest (RF) model in predicting the poor prognosis of SFTS patients. Additionally, we validated the predictive effects of MCP-3 through receiver operating characteristic (ROC) curve analysis with an AUC of 0.882 (95% CI, 0.787-0.978, P <0.001), and the clinical applicability of MCP-3 was assessed favorably based on decision curve analysis (DCA). The Spearman correlation analysis indicated that the level of MCP-3 was positively correlated with ALT, AST, LDH, α-HBDH, APTT, D-dimer, and viral load (P<0.01). Discussion For the first time, our study identified and validated that MCP-3 could serve as a meaningful biomarker for predicting the fatal outcome of SFTS patients. The longitudinal cytokine profile analyzed that abnormally increased cytokines were associated with the poor prognosis of SFTS patients. Our study provides new insights into exploring the pathogenesis of cytokines with organ damage and leading to adverse effects.
Collapse
Affiliation(s)
- Zishuai Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Chenxi Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Hong Yu
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, Shandong, China
| | - Rongling Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xiaoyu Xue
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhouling Jiang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ziruo Ge
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yanli Xu
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, Shandong, China
| | - Wei Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Ling Lin
- Department of Infectious Diseases, Yantai Qishan Hospital, Yantai, Shandong, China
| | - Zhihai Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Hurme P, Sahla R, Rückert B, Vahlberg T, Turunen R, Vuorinen T, Akdis M, Söderlund‐Venermo M, Akdis C, Jartti T. Human bocavirus 1 coinfection is associated with decreased cytokine expression in the rhinovirus-induced first wheezing episode in children. Clin Transl Allergy 2023; 13:e12311. [PMID: 38006383 PMCID: PMC10642552 DOI: 10.1002/clt2.12311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/20/2023] [Accepted: 10/22/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Rhinovirus (RV)-induced first wheezing episodes in children are associated with a markedly increased risk of asthma. Previous studies have suggested that human bocavirus 1 (HBoV1) may modify RV-induced immune responses in young children. We investigated cytokine profiles of sole RV- and dual RV-HBoV1-induced first wheezing episodes, and their association with severity and prognosis. METHODS Fifty-two children infected with only RV and nine children infected with dual RV-HBoV1, aged 3-23 months, with severe first wheezing episodes were recruited. At acute illness and 2 weeks later, peripheral blood mononuclear cells were isolated, and stimulated with anti-CD3/anti-CD28 in vitro. Multiplex ELISA was used to quantitatively identify 56 different cytokines at both study points. Patients were prospectively followed for 4 years. RESULTS The mean age of the children was 14.3 months, and 30% were sensitized. During the acute illness, the adjusted analyses revealed a decrease in the expression of IL-1b, MIP-1b, Regulated upon activation, normal T cell expressed and presumably secreted (CCL5), TNF-a, TARC, and ENA-78 in the RV-HBoV1 group compared with the RV group. In the convalescence phase, the RV-HBoV1 group was characterized by decreased expression of Fractalkine, MCP-3, and IL-8 compared to the RV group. Furthermore, the hospitalization time was associated with the virus group and cytokine response (interaction p < 0.05), signifying that increased levels of epidermal growth factor and MIP-1b were related with a shorter duration of hospitalization in the RV-HBoV1 coinfection group but not in the RV group. CONCLUSIONS Different cytokine response profiles were detected between the RV and the RV-HBoV1 groups. Our results show the idea that RV-induced immune responses may be suppressed by HBoV1.
Collapse
Affiliation(s)
- Pekka Hurme
- Department of Pediatrics and Adolescent MedicineTurku University HospitalUniversity of TurkuTurkuFinland
| | - Reetta Sahla
- Department of Pediatrics and Adolescent MedicineTurku University HospitalUniversity of TurkuTurkuFinland
| | - Beate Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZürichChristine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - Tero Vahlberg
- Department of BiostatisticsUniversity of TurkuTurkuFinland
| | - Riitta Turunen
- Department of Pediatrics and Adolescent MedicineTurku University HospitalUniversity of TurkuTurkuFinland
- New Children's HospitalHelsinki University HospitalUniversity of HelsinkiHelsinkiFinland
| | - Tytti Vuorinen
- Institute of BiomedicineUniversity of TurkuTurkuFinland
- Department of Clinical MicrobiologyTurku University HospitalTurkuFinland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZürichChristine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | | | - Cezmi Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZürichChristine Kühne‐Center for Allergy Research and Education (CK‐CARE)DavosSwitzerland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent MedicineTurku University HospitalUniversity of TurkuTurkuFinland
- Research Unit of Clinical MedicineMedical Research CenterUniversity of OuluOuluFinland
- Department of Pediatrics and Adolescent MedicineOulu University HospitalOuluFinland
| |
Collapse
|
3
|
Li L, Dai F, Wang L, Sun Y, Mei L, Ran Y, Ye F. CCL13 and human diseases. Front Immunol 2023; 14:1176639. [PMID: 37153575 PMCID: PMC10154514 DOI: 10.3389/fimmu.2023.1176639] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/05/2023] [Indexed: 05/09/2023] Open
Abstract
CCL13/MCP-4 belongs to the CC chemokine family, which induces chemotaxis in many immune cells. Despite extensive research into its function in numerous disorders, a thorough analysis of CCL13 is not yet accessible. The role of CCL13 in human disorders and existing CCL13-focused therapies are outlined in this study. The function of CCL13 in rheumatic diseases, skin conditions, and cancer is comparatively well-established, and some studies also suggest that it may be involved in ocular disorders, orthopedic conditions, nasal polyps, and obesity. We also give an overview of research that found very little evidence of CCL13 in HIV, nephritis, and multiple sclerosis. Even though CCL13-mediated inflammation is frequently linked to disease pathogenesis, it's fascinating to note that in some conditions, like primary biliary cholangitis (PBC) and suicide, it might even act as a preventative measure.
Collapse
Affiliation(s)
- Laifu Li
- Department of Gastroenterology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Province Key Laboratory of Gastrointestinal Motility Disorders, Laboratory of Digestive Diseases of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, China
| | - Fei Dai
- Department of Gastroenterology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Province Key Laboratory of Gastrointestinal Motility Disorders, Laboratory of Digestive Diseases of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, China
- *Correspondence: Fei Dai,
| | - Lianli Wang
- Department of Gastroenterology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Province Key Laboratory of Gastrointestinal Motility Disorders, Laboratory of Digestive Diseases of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, China
| | - Yating Sun
- Department of Gastroenterology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Province Key Laboratory of Gastrointestinal Motility Disorders, Laboratory of Digestive Diseases of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, China
| | - Lin Mei
- Department of Gastroenterology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Province Key Laboratory of Gastrointestinal Motility Disorders, Laboratory of Digestive Diseases of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, China
| | - Yan Ran
- Department of Gastroenterology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Province Key Laboratory of Gastrointestinal Motility Disorders, Laboratory of Digestive Diseases of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, China
| | - Fangchen Ye
- Department of Gastroenterology, Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
- Shaanxi Province Key Laboratory of Gastrointestinal Motility Disorders, Laboratory of Digestive Diseases of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi’an, China
| |
Collapse
|
4
|
She S, Ren L, Chen P, Wang M, Chen D, Wang Y, Chen H. Functional Roles of Chemokine Receptor CCR2 and Its Ligands in Liver Disease. Front Immunol 2022; 13:812431. [PMID: 35281057 PMCID: PMC8913720 DOI: 10.3389/fimmu.2022.812431] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Chemokines are a family of cytokines that orchestrate the migration and positioning of immune cells within tissues and are critical for the function of the immune system. CCR2 participates in liver pathology, including acute liver injury, chronic hepatitis, fibrosis/cirrhosis, and tumor progression, by mediating the recruitment of immune cells to inflammation and tumor sites. Although a variety of chemokines have been well studied in various diseases, there is no comprehensive review presenting the roles of all known chemokine ligands of CCR2 (CCL2, CCL7, CCL8, CCL12, CCL13, CCL16, and PSMP) in liver disease, and this review aims to fill this gap. The introduction of each chemokine includes its discovery, its corresponding chemotactic receptors, physiological functions and roles in inflammation and tumors, and its impact on different immune cell subgroups.
Collapse
Affiliation(s)
- Shaoping She
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People’s Hospital, Beijing, China
| | - Liying Ren
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Pu Chen
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People’s Hospital, Beijing, China
| | - Mingyang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Dongbo Chen
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People’s Hospital, Beijing, China
| | - Ying Wang
- Department of Immunology, School of Basic Medical Sciences, and NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Hongsong Chen
- Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Peking University People’s Hospital, Beijing, China
- *Correspondence: Hongsong Chen,
| |
Collapse
|
5
|
Priyadharshini V, Jiménez-Chobillon MA, de Graaf J, Porras Gutiérrez de Velasco R, Gratziou C, Ramírez-Jiménez F, Teran LM. Transcriptome Analysis Identifies Doublesex and Mab-3 Related Transcription Factor (DMRT3) in Nasal Polyp Epithelial Cells of Patients Suffering from Non-Steroidal Anti-Inflammatory Drug-Exacerbated Respiratory Disease (AERD). Biomolecules 2021; 11:biom11081092. [PMID: 34439758 PMCID: PMC8394795 DOI: 10.3390/biom11081092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/17/2021] [Accepted: 07/19/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Aspirin-exacerbated respiratory disease (AERD) is a syndrome characterised by chronic rhinosinusitis, nasal polyps, asthma and aspirin intolerance. An imbalance of eicosanoid metabolism with anover-production of cysteinyl leukotrienes (CysLTs) has been associated with AERD. However, the precise mechanisms underlying AERD are unknown. Objective: To establish the transcriptome of the nasal polyp airway epithelial cells derived from AERD patients to discover gene expression patterns in this disease. Methods: Nasal airway epithelial cells were isolated from 12 AERD polyps and 8 AERD non-polyp nasal mucosa samples as controls from the same subjects. Utilising the Illumina HiSeq 2500 platform, RNA samples were sequenced. Potential gene candidate DMRT3 was selected from the differentially-expressed genes for validation. Results: Comparative transcriptome profiling of nasal epithelial cells was accomplished in AERD. A total of 20 genes had twofold mean regulation expression differences or greater. In addition, 8 genes were upregulated, including doublesex and mab-3 related transcription factor 3 (DMRT3), and 12 genes were downregulated. Differentially regulated genes comprised roles in inflammation, defence and immunity. Metabolic process and embryonic development pathways were significantly enriched. Enzyme-linked immune sorbent assay (ELISA) results of DMRT3 in AERD patients were significantly upregulated compared to controls (p = 0.03). Immunohistochemistry (IHC) of AERD nasal polyps localised DMRT3 and was predominantly released in the airway epithelia. Conclusion: Findings suggest that DMRT3 could be potentially involved in nasal polyp development in AERD patients. Furthermore, several genes are downregulated, hinting at the dedifferentiation phenomenon in AERD polyps. However, further studies are imperative to confirm the exact mechanism of polyp formation in AERD patients.
Collapse
Affiliation(s)
- V.S. Priyadharshini
- Instituto Nacional de EnfermedadesRespiratorias Ismael Cosío Villegas, Calz. de Tlalpan 4502, Belisario Domínguez Secc 16, Mexico City 14080, Mexico; (V.S.P.); (M.A.J.-C.); (F.R.-J.)
| | - Marcos Alejandro Jiménez-Chobillon
- Instituto Nacional de EnfermedadesRespiratorias Ismael Cosío Villegas, Calz. de Tlalpan 4502, Belisario Domínguez Secc 16, Mexico City 14080, Mexico; (V.S.P.); (M.A.J.-C.); (F.R.-J.)
| | - Jos de Graaf
- Translational Oncology at Johannes Gutenberg-University Medical Center gGmbH, D-55131 Mainz, Germany;
| | - Raúl Porras Gutiérrez de Velasco
- School of Medicine, Universidad Nacional Autónoma de México, Av. Universidad 3000, Circuito Exterior S/N. Delegación Coyoacán, Mexico City 04510, Mexico;
| | - Christina Gratziou
- Smoking Cessation Centre Pulmonary Department, Evgenidio Hospital, Athens University, 20 Papadiamantopoulou Street, 11528 Athens, Greece;
| | - Fernando Ramírez-Jiménez
- Instituto Nacional de EnfermedadesRespiratorias Ismael Cosío Villegas, Calz. de Tlalpan 4502, Belisario Domínguez Secc 16, Mexico City 14080, Mexico; (V.S.P.); (M.A.J.-C.); (F.R.-J.)
| | - Luis M. Teran
- Instituto Nacional de EnfermedadesRespiratorias Ismael Cosío Villegas, Calz. de Tlalpan 4502, Belisario Domínguez Secc 16, Mexico City 14080, Mexico; (V.S.P.); (M.A.J.-C.); (F.R.-J.)
- School of Medicine, Universidad Nacional Autónoma de México, Av. Universidad 3000, Circuito Exterior S/N. Delegación Coyoacán, Mexico City 04510, Mexico;
- Correspondence:
| |
Collapse
|
6
|
Lai Y, Zhou X, Guo F, Jin X, Meng G, Zhou L, Chen H, Liu Z, Yu L, Jiang H. Non-invasive transcutaneous vagal nerve stimulation improves myocardial performance in doxorubicin-induced cardiotoxicity. Cardiovasc Res 2021; 118:1821-1834. [PMID: 34145895 DOI: 10.1093/cvr/cvab209] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 06/17/2021] [Indexed: 12/29/2022] Open
Abstract
AIMS The clinical use of antitumor agent doxorubicin (DOX) is hampered by its dose-dependent cardiotoxicity. Development of highly efficient and safe adjuvant intervention for preventing DOX-induced adverse cardiac events is urgently needed. We aimed to investigate whether transcutaneous vagal nerve stimulation (tVNS) plays a cardio-protective role in DOX-induced cardiotoxicity. METHODS AND RESULTS Healthy male adult Sprague Dawley rats were used in the experiment and were randomly divided into four groups including control, DOX, tVNS and DOX+tVNS groups. A cumulative dose of 15 mg/kg DOX was intraperitoneally injected into rats to generate cardiotoxicity. Non-invasive tVNS was conducted for 6 weeks (30 min/day). After six-week intervention, the indices from the echocardiography revealed that tVNS significantly improved left ventricular function compared to the DOX group. The increased malondialdehyde (MDA) and Interleukin-1β (IL-1β), and decreased superoxide dismutase (SOD) were observed in the DOX group, while tVNS significantly prevented these changes. From cardiac histopathological analysis, the DOX+tVNS group showed a mild myocardial damage, and decreases in cardiac fibrosis and myocardial apoptosis compared to the DOX group. Heart rate variability (HRV) analysis showed that tVNS significantly inhibited DOX-induced sympathetic hyperactivity compared to the DOX group. Additionally, the results of RNA-sequencing analysis showed that there were 245 differentially expressed genes in the DOX group compared to the control group, among which 39 genes were downregulated by tVNS and most of these genes were involved in immune system. Moreover, tVNS significantly downregulated the relative mRNA expressions of chemokine-related genes and macrophages recruitment compared to the DOX group. CONCLUSION These results suggest that tVNS prevented DOX-induced cardiotoxicity by rebalancing autonomic tone, ameliorating cardiac dysfunction and remodeling. Notably, crosstalk between autonomic neuromodulation and innate immune cells macrophages mediated by chemokines might be involved in the underlying mechanisms. A TRANSLATIONAL PERSPECTIVE Non-invasive tVNS has been identified an effective neuromodulation strategy exerting beneficial effects on rebalancing autonomic tone and cardiac pathological conditions. The present study provided direct evidence for a beneficial role of tVNS in preventing DOX-induced autonomic dysfunction and cardiotoxicity in vivo. Additionally, recent studies revealed the importance of sympathetic nerve fibers involving in tumorigenesis and the benefits of higher vagal tone for tumor prognosis either in animal or human trials. Together, tVNS may not only become a novel, nonpharmacological adjuvant therapy for preventing doxorubicin-induced cardiotoxicity, but also may be beneficial for prognosis of cancer patients during chemotherapy. In our future study, we would investigate the effect of tVNS on both combined chemotherapy-induced cardiotoxicity and the antitumor efficacy of DOX in tumor models.
Collapse
Affiliation(s)
- Yanqiu Lai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Fuding Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Xiaoxing Jin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Guannan Meng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Hu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
- Cardiac Autonomic Nervous System Research Center of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan City, Hubei Province, 430060, China
| |
Collapse
|
7
|
Lewis TC, Metitiri EE, Mentz GB, Ren X, Carpenter AR, Goldsmith AM, Wicklund KE, Eder BN, Comstock AT, Ricci JM, Brennan SR, Washington GL, Owens KB, Mukherjee B, Robins TG, Batterman SA, Hershenson MB. Influence of viral infection on the relationships between airway cytokines and lung function in asthmatic children. Respir Res 2018; 19:228. [PMID: 30463560 PMCID: PMC6249926 DOI: 10.1186/s12931-018-0922-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/24/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Few longitudinal studies examine inflammation and lung function in asthma. We sought to determine the cytokines that reduce airflow, and the influence of respiratory viral infections on these relationships. METHODS Children underwent home collections of nasal lavage during scheduled surveillance periods and self-reported respiratory illnesses. We studied 53 children for one year, analyzing 392 surveillance samples and 203 samples from 85 respiratory illnesses. Generalized estimated equations were used to evaluate associations between nasal lavage biomarkers (7 mRNAs, 10 proteins), lung function and viral infection. RESULTS As anticipated, viral infection was associated with increased cytokines and reduced FVC and FEV1. However, we found frequent and strong interactions between biomarkers and virus on lung function. For example, in the absence of viral infection, CXCL10 mRNA, MDA5 mRNA, CXCL10, IL-4, IL-13, CCL4, CCL5, CCL20 and CCL24 were negatively associated with FVC. In contrast, during infection, the opposite relationship was frequently found, with IL-4, IL-13, CCL5, CCL20 and CCL24 levels associated with less severe reductions in both FVC and FEV1. CONCLUSIONS In asthmatic children, airflow obstruction is driven by specific pro-inflammatory cytokines. In the absence of viral infection, higher cytokine levels are associated with decreasing lung function. However, with infection, there is a reversal in this relationship, with cytokine abundance associated with reduced lung function decline. While nasal samples may not reflect lower airway responses, these data suggest that some aspects of the inflammatory response may be protective against viral infection. This study may have ramifications for the treatment of viral-induced asthma exacerbations.
Collapse
Affiliation(s)
- Toby C. Lewis
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
- Health Behavior/Health Education, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Ediri E. Metitiri
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Graciela B. Mentz
- Health Behavior/Health Education, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Xiaodan Ren
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Ashley R. Carpenter
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Adam M. Goldsmith
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Kyra E. Wicklund
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
- Epidemiology, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Breanna N. Eder
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Adam T. Comstock
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Jeannette M. Ricci
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Sean R. Brennan
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Ginger L. Washington
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Kendall B. Owens
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
| | - Bhramar Mukherjee
- Departments of Biostatistics, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Thomas G. Robins
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Stuart A. Batterman
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| | - Marc B. Hershenson
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
- Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, USA
| | - the Community Action Against Asthma Steering Committee
- Departments of Pediatrics and Communicable Diseases, University of Michigan Medical School, 1150 W. Medical Center Dr., Building MSRB2, Room 3570B, Ann Arbor, MI 48109-5688 USA
- Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, USA
- Departments of Biostatistics, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
- Environmental Health Sciences, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
- Epidemiology, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
- Health Behavior/Health Education, University of Michigan School of Public Health, University of Michigan, Ann Arbor, MI 48109 USA
| |
Collapse
|
8
|
Lewis TC, Metitiri EE, Mentz GB, Ren X, Goldsmith AM, Eder BN, Wicklund KE, Walsh MP, Comstock AT, Ricci JM, Brennan SR, Washington GL, Owens KB, Mukherjee B, Robins TG, Batterman SA, Hershenson MB. Impact of community respiratory viral infections in urban children with asthma. Ann Allergy Asthma Immunol 2018; 122:175-183.e2. [PMID: 30385348 PMCID: PMC6360098 DOI: 10.1016/j.anai.2018.10.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/13/2018] [Accepted: 10/21/2018] [Indexed: 12/13/2022]
Abstract
Background Upper respiratory tract viral infections cause asthma exacerbations in children. However, the impact of natural colds on children with asthma in the community, particularly in the high-risk urban environment, is less well defined. Objective We hypothesized that children with high-symptom upper respiratory viral infections have reduced airway function and greater respiratory tract inflammation than children with virus-positive low-symptom illnesses or virus-negative upper respiratory tract symptoms. Methods We studied 53 children with asthma from Detroit, Michigan, during scheduled surveillance periods and self-reported respiratory illnesses for 1 year. Symptom score, spirometry, fraction of exhaled nitric oxide (FeNO), and nasal aspirate biomarkers, and viral nucleic acid and rhinovirus (RV) copy number were assessed. Results Of 658 aspirates collected, 22.9% of surveillance samples and 33.7% of respiratory illnesses were virus-positive. Compared with the virus-negative asymptomatic condition, children with severe colds (symptom score ≥5) showed reduced forced expiratory flow at 25% to 75% of the pulmonary volume (FEF25%-75%), higher nasal messenger RNA expression of C-X-C motif chemokine ligand (CXCL)-10 and melanoma differentiation-associated protein 5, and higher protein abundance of CXCL8, CXCL10 and C-C motif chemokine ligands (CCL)-2, CCL4, CCL20, and CCL24. Children with mild (symptom score, 1-4) and asymptomatic infections showed normal airway function and fewer biomarker elevations. Virus-negative cold-like illnesses demonstrated increased FeNO, minimal biomarker elevation, and normal airflow. The RV copy number was associated with nasal chemokine levels but not symptom score. Conclusion Urban children with asthma with high-symptom respiratory viral infections have reduced FEF25%-75% and more elevations of nasal biomarkers than children with mild or symptomatic infections, or virus-negative illnesses.
Collapse
Affiliation(s)
- Toby C Lewis
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan; Department of Environmental Health Sciences, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan; Department of Health Behavior/Health Education, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Ediri E Metitiri
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Graciela B Mentz
- Department of Health Behavior/Health Education, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Xiaodan Ren
- Department of Environmental Health Sciences, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Adam M Goldsmith
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Breanna N Eder
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kyra E Wicklund
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan; Department of Epidemiology, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Megan P Walsh
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan; Department of Epidemiology, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Adam T Comstock
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jeannette M Ricci
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Sean R Brennan
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Ginger L Washington
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Kendall B Owens
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Bhramar Mukherjee
- Department of Biostatistics, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Thomas G Robins
- Department of Environmental Health Sciences, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Stuart A Batterman
- Department of Environmental Health Sciences, University of Michigan School of Public Health; University of Michigan, Ann Arbor, Michigan
| | - Marc B Hershenson
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan; Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan.
| | | |
Collapse
|
9
|
Human Neutrophil Defensin-1, -3, and -4 Are Elevated in Nasal Aspirates from Children with Naturally Occurring Adenovirus Infection. Can Respir J 2018; 2018:1038593. [PMID: 30154940 PMCID: PMC6091448 DOI: 10.1155/2018/1038593] [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: 12/06/2017] [Revised: 04/17/2018] [Accepted: 07/04/2018] [Indexed: 12/26/2022] Open
Abstract
Background Adenoviruses are highly contagious pathogens which cause respiratory disease particularly in children; they may induce severe disease in infants. Human neutrophil peptides (HNPs) have been found to exhibit antiadenoviral activity. Thus, we have investigated HNPs in nasal aspirates (NAs) of children suffering from adenoviral common cold. Objective To investigate the release of HNP-1–4 in adenovirus infection and the relationship with self-limiting upper respiratory tract infections. Methods Nasal aspirate samples (n=14) were obtained from children (aged 6–12 years) infected with adenovirus between June 2012 and December 2015. Control samples were taken 4 weeks after infection when the children were asymptomatic. Levels of HNPs were measured using an enzyme-linked immunosorbent assay (ELISA). Results There were increased levels of HNP-1, -3, and -4, but not HNP-2, in nasal aspirates (NAs) during adenovirus infections compared to healthy specimens (p ≤ 0.01). Moreover, there was also increase in the neutrophil count, which is a known cell source of HNPs. Conclusion Our finding supports the involvement of HNP-1, -3, and -4 in naturally occurring cold in children infected with adenovirus. Because of their known antiviral properties, it is tempting to hypothesize that HNPs might play a protective role in adenovirus-induced respiratory disease; however, this remains to be shown.
Collapse
|
10
|
Girkin J, Hatchwell L, Foster P, Johnston SL, Bartlett N, Collison A, Mattes J. CCL7 and IRF-7 Mediate Hallmark Inflammatory and IFN Responses following Rhinovirus 1B Infection. THE JOURNAL OF IMMUNOLOGY 2015; 194:4924-30. [PMID: 25847975 DOI: 10.4049/jimmunol.1401362] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 03/13/2015] [Indexed: 12/22/2022]
Abstract
Rhinovirus (RV) infections are common and have the potential to exacerbate asthma. We have determined the lung transcriptome in RV strain 1B-infected naive BALB/c mice (nonallergic) and identified CCL7 and IFN regulatory factor (IRF)-7 among the most upregulated mRNA transcripts in the lung. To investigate their roles we employed anti-CCL7 Abs and an IRF-7-targeting small interfering RNA in vivo. Neutralizing CCL7 or inhibiting IRF-7 limited neutrophil and macrophage influx and IFN responses in nonallergic mice. Neutralizing CCL7 also reduced activation of NF-κB p65 and p50 subunits, as well as airway hyperreactivity (AHR) in nonallergic mice. However, neither NF-κB subunit activation nor AHR was abolished with infection of allergic mice after neutralizing CCL7, despite a reduction in the number of neutrophils, macrophages, and eosinophils. IRF-7 small interfering RNA primarily suppressed IFN-α and IFN-β levels during infection of allergic mice. Our data highlight a pivotal role of CCL7 and IRF-7 in RV-induced inflammation and IFN responses and link NF-κB signaling to the development of AHR.
Collapse
Affiliation(s)
- Jason Girkin
- Experimental and Translational Respiratory Medicine Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2305, Australia; Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia
| | - Luke Hatchwell
- Experimental and Translational Respiratory Medicine Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2305, Australia; Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia
| | - Paul Foster
- Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia
| | - Sebastian L Johnston
- Airway Disease Infection Section, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London W2 1PG, United Kingdom; and
| | - Nathan Bartlett
- Airway Disease Infection Section, National Heart and Lung Institute, Medical Research Council and Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London W2 1PG, United Kingdom; and
| | - Adam Collison
- Experimental and Translational Respiratory Medicine Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2305, Australia; Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia
| | - Joerg Mattes
- Experimental and Translational Respiratory Medicine Group, University of Newcastle and Hunter Medical Research Institute, Newcastle, New South Wales 2305, Australia; Priority Research Centre for Asthma and Respiratory Diseases, University of Newcastle, Newcastle, New South Wales 2305, Australia; Paediatric Respiratory and Sleep Medicine Unit, Newcastle Children's Hospital, Kaleidoscope, Newcastle, New South Wales 2305, Australia
| |
Collapse
|
11
|
Zanotta N, Maximova N, Campisciano G, Del Savio R, Pizzol A, Casalicchio G, Berton E, Comar M. Up-regulation of the monocyte chemotactic protein-3 in sera from bone marrow transplanted children with torquetenovirus infection. J Clin Virol 2014; 63:6-11. [PMID: 25600596 DOI: 10.1016/j.jcv.2014.11.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/06/2014] [Accepted: 11/22/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND Torquetenovirus (TTV) represents a commensal human virus producing life-long viremia in approximately 80% of healthy individuals of all ages. A potential pathogenic role for TTV has been suggested in immunocompromised patients with hepatitis of unknown etiology sustained by strong proinflammatory cytokines. OBJECTIVES The aim of this study was to investigate the sera immunological profile linked to TTV infection in bone marrow transplant (BMT) children with liver injury. STUDY DESIGN TTV infection was assessed in sera from 27 BMT patients with altered hepatic parameters and histological features, by the use of quantitative real-time PCR, along with TTV genogroups and coinfection with HEV. The qualitative and quantitative nature of soluble inflammatory factors was evaluated studying a large set of cytokines using the Bioplex platform. As controls, sera from 22 healthy children negative for serological and molecular hepatitis markers including TTV and HEV, and for autoimmune diseases, were selected. RESULTS AND CONCLUSIONS TTV was detected in 81.4% of BMT patients with a viral load ranging from 10(5) to 10(9) copies/mL. All samples were HEV-RNA negative. A pattern of cytokines, IFN-γ, TNF-α, FGF-basic (p<0.01) and MCP-3 (p<0.001) was found significantly highly expressed in TTV-positive patients compared to TTV-negative and controls. Of note, MCP-3 chemokine showed the highest sera concentration independently of the amount of TTV load and the status of immune system deregulation (p<0.001). In this pilot study for the first time, a positive association was found between TTV and increased level of MCP-3 suggesting a indirect role of TTV in liver injury.
Collapse
Affiliation(s)
- Nunzia Zanotta
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - Natalia Maximova
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | | | - Rossella Del Savio
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - Antonio Pizzol
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - Giorgia Casalicchio
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - Emanuela Berton
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - Manola Comar
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy; Department of Medical Science, University of Trieste, Trieste, Italy.
| |
Collapse
|
12
|
Teran LM, Montes-Vizuet R, Li X, Franz T. Respiratory proteomics: from descriptive studies to personalized medicine. J Proteome Res 2014; 14:38-50. [PMID: 25382407 DOI: 10.1021/pr500935s] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Respiratory diseases are highly prevalent and affect humankind worldwide, causing extensive morbidity and mortality with the environment playing an important role. Given the complex structure of the airways, sophisticated tools are required for early diagnosis; initial symptoms are nonspecific, and the clinical diagnosis is made frequently late. Over the past few years, proteomics has made high technological progress in mass-spectrometry-based protein identification and has allowed us to gain new insights into disease mechanisms and identify potential novel therapeutic targets. This review will highlight the contributions of proteomics toward the understanding of the respiratory proteome listing potential biomarkers and its potential application to the clinic. We also outline the contributions of proteomics to creating a personalized approach in respiratory medicine.
Collapse
Affiliation(s)
- Luis M Teran
- Instituto Nacional de Enfermedades Respiratorias , Calz. de Tlalpan 4502, Distrito Federal 14080, Mexico
| | | | | | | |
Collapse
|
13
|
Cheng JW, Sadeghi Z, Levine AD, Penn MS, von Recum HA, Caplan AI, Hijaz A. The role of CXCL12 and CCL7 chemokines in immune regulation, embryonic development, and tissue regeneration. Cytokine 2014; 69:277-83. [PMID: 25034237 DOI: 10.1016/j.cyto.2014.06.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Revised: 05/23/2014] [Accepted: 06/04/2014] [Indexed: 12/20/2022]
Abstract
Chemotactic factors direct the migration of immune cells, multipotent stem cells, and progenitor cells under physiologic and pathologic conditions. Chemokine ligand 12 and chemokine ligand 7 have been identified and investigated in multiple studies for their role in cellular trafficking in the setting of tissue regeneration. Recent early phase clinical trials have suggested that these molecules may lead to clinical benefit in patients with chronic disease. Importantly, these two proteins may play additional significant roles in directing the migration of multipotent cells, such as mesenchymal stem cells and hematopoietic progenitor cells. This article reviews the functions of these two chemokines, focusing on recruitment to sites of injury, immune function modulation, and contributions to embryonic development. Additional research would provide valuable insight into the potential clinical application of these two proteins in stem cell therapy.
Collapse
Affiliation(s)
- Julie W Cheng
- Urology Institute, University Hospitals Case Medical Center, Department of Urology, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, United States
| | - Zhina Sadeghi
- Urology Institute, University Hospitals Case Medical Center, Department of Urology, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, United States
| | - Alan D Levine
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States; Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, United States
| | - Marc S Penn
- Skirball Laboratory for Cardiovascular Cellular Therapeutics, Summa Cardiovascular Institute, Summa Health System, 525 East Market Street, Akron, OH 44304, United States
| | - Horst A von Recum
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States
| | - Arnold I Caplan
- Skeletal Research Center, Department of Biology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, United States
| | - Adonis Hijaz
- Urology Institute, University Hospitals Case Medical Center, Department of Urology, Case Western Reserve University School of Medicine, 11100 Euclid Avenue, Cleveland, OH 44106, United States.
| |
Collapse
|
14
|
Lee JH, Park CS. Gene - Gene Interactions Among MCP Genes Polymorphisms in Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2014; 6:333-40. [PMID: 24991457 PMCID: PMC4077960 DOI: 10.4168/aair.2014.6.4.333] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 11/19/2013] [Accepted: 11/27/2013] [Indexed: 11/20/2022]
Abstract
Purpose Monocyte chemoattractant proteins (MCPs) are important cytokines that involved in cellular activation and releasing of inflammatoy mediators by basophils and eosinophils in allergic disease. Some MCP gene variants implicate in asthma and monoclonal antibody for MCP-3 blocks allergic inflammations in the patients with asthma. Detection of interactions between gene and environment or between genes for complex disease such as asthma is important. We searched for an evidence of genetic effect of single nucleotide polymorphisms (SNPs) of MCP genes as well as gene - gene interactions involved in asthma. Methods Four hundreds asthmatics and four hundreds normal controls were enrolled. Asthma was defined as a positive bronchodilator response or positive methacholine provocation test with compatible clinical symptoms. Seven MCP gene SNPs (2 SNPs in MCP-1, 1 in MCP-2, and 4 in MCP-3) were included. Association analyses between SNP and asthma, and the tests for gene - gene interaction were performed. Results Strong linkage disequilibria were found among 7 MCP gene polymorphisms. There was no SNP that showed a significant association with asthma among 7 SNPs of 3 MCP genes. No haplotype was associated with asthma, either. The combination of MCP1-2518G>A, MCP2+46A>C, and MCP3+563C>T was the best predictive model for asthma as compared to the control in tests for gene - gene interaction. The MCP1-2518G>A and MCP2+46A>C was the second best predictive combination and this had the highest synergistic interaction effect on the subject's status than any other combination of polymorphisms. Complete linkages were not associated with the gene - gene interactions models. Conclusions MCP gene polymorphisms probably interact with each other; thus, these findings may help in developing a possible genetic marker to predict asthma.
Collapse
Affiliation(s)
- June-Hyuk Lee
- Respiratory and Allergy Medicine, Interanl Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Choon-Sik Park
- Respiratory and Allergy Medicine, Interanl Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| |
Collapse
|
15
|
Asthma exacerbations: predisposing factors and prediction rules. Curr Opin Allergy Clin Immunol 2014; 13:225-36. [PMID: 23635528 DOI: 10.1097/aci.0b013e32836096de] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Asthma is a multifaceted disease that is associated with decreased lung function, multiple symptoms, varying levels of asthma control, and risk of acute exacerbations. The ability to predict the risk of developing acute exacerbations may improve the management of asthmatics and facilitate identification of these patients for interventional studies. RECENT FINDINGS Factors that are associated with different manifestations of asthma differ. Biomarkers that are correlated with airways hyper-responsiveness do not necessarily correlate with risk of future exacerbations. Genetic factors that segregate with exacerbation risk are beginning to emerge. Outcome measures that demonstrate predictive validity have been developed and may facilitate patient management and provide novel clinically meaningful endpoints in clinical trials. SUMMARY This review will emphasize underlying factors associated with asthma exacerbations and clinical prediction rules that correlate with the risk of developing severe exacerbations of asthma.
Collapse
|
16
|
Sadakane K, Ichinose T, Takano H, Yanagisawa R, Koike E, Inoue KI. The alkylphenols 4-nonylphenol, 4-tert-octylphenol and 4-tert-butylphenol aggravate atopic dermatitis-like skin lesions in NC/Nga mice. J Appl Toxicol 2013; 34:893-902. [DOI: 10.1002/jat.2911] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 06/21/2013] [Accepted: 06/21/2013] [Indexed: 11/09/2022]
Affiliation(s)
- Kaori Sadakane
- Department of Health Sciences; Oita University of Nursing and Health Sciences; Megusuno Oita City Oita Japan
| | - Takamichi Ichinose
- Department of Health Sciences; Oita University of Nursing and Health Sciences; Megusuno Oita City Oita Japan
| | - Hirohisa Takano
- Department of Environmental Engineering; Kyoto University Graduate School of Engineering; Nishikyo-ku Kyoto Japan
| | - Rie Yanagisawa
- Center for Environmental Health Sciences; National Institute for Environmental Studies; Tsukuba City Ibaraki Japan
| | - Eiko Koike
- Center for Environmental Health Sciences; National Institute for Environmental Studies; Tsukuba City Ibaraki Japan
| | - Ken-ichiro Inoue
- Center for Medical Science; International University of Health and Welfare; Otawara Tochigi Japan
| |
Collapse
|
17
|
Mendez-Enriquez E, García-Zepeda EA. The multiple faces of CCL13 in immunity and inflammation. Inflammopharmacology 2013; 21:397-406. [PMID: 23846739 DOI: 10.1007/s10787-013-0177-5] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/20/2013] [Indexed: 01/19/2023]
Abstract
CCL13/MCP-4, is a CC family chemokine that is chemoattractant for eosinophils, basophils, monocytes, macrophages, immature dendritic cells, and T cells, and its capable of inducing crucial immuno-modulatory responses through its effects on epithelial, muscular and endothelial cells. Similar to other CC chemokines, CCL13 binds to several chemokine receptors (CCR1, CCR2 and CCR3), allowing it to elicit different effects on its target cells. A number of studies have shown that CCL13 is involved in many chronic inflammatory diseases, in which it functions as a pivotal molecule involved in the selective recruitment of cell lineages to the inflamed tissues and their subsequent activation. Based on these studies, we suggest that blocking the actions of CCL13 can serve as a novel strategy for the generation of agents with anti-inflammatory activity. The main goal of this review is to present the current information about CCL13, its gene and protein structure and the roles of this chemokine during innate/adaptive immune responses in inflammatory diseases.
Collapse
Affiliation(s)
- E Mendez-Enriquez
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar S/N, Ciudad Universitaria, 04510, México, DF, México
| | | |
Collapse
|
18
|
Lewis TC, Henderson TA, Carpenter AR, Ramirez IA, McHenry CL, Goldsmith AM, Ren X, Mentz GB, Mukherjee B, Robins TG, Joiner TA, Mohammad LS, Nguyen ER, Burns MA, Burke DT, Hershenson MB. Nasal cytokine responses to natural colds in asthmatic children. Clin Exp Allergy 2013. [PMID: 23181789 PMCID: PMC4219353 DOI: 10.1111/cea.12005] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The mechanisms by which viruses induce asthma exacerbations are not well understood. Objective We characterized fluctuations in nasal aspirate cytokines during naturally occurring respiratory viral infections in children with asthma. Methods Sixteen children underwent home collections of nasal aspirates when they were without cold symptoms and again during self‐reported respiratory illnesses. The presence of viral infection was ascertained by multiplex PCR. Cytokines were measured using multiplex immune assay. mRNA expression for selected markers of viral infection was measured using RT‐PCR. A cumulative respiratory symptom score was calculated for each day of measurement. Generalized estimated equations were used to evaluate associations between viral infection and marker elevation, and between marker elevation and symptom score. Results The 16 patients completed a total of 37 weeks of assessment (15 ‘well’ weeks; 22 self‐assessed ‘sick’ weeks). Viral infections were detected in 3 of the ‘well’ weeks and 17 of the ‘sick’ weeks (10 rhinovirus, three coronavirus, two influenza A, two influenza B, two respiratory syncytial virus, one parainfluenza). Compared to virus‐negative well weeks, nasal aspirate IFN‐γ, CXCL8/IL‐8, CXCL10/IP‐10, CCL5/RANTES, CCL11/eotaxin‐1, CCL2/MCP‐1, CCL4/MIP‐1β, CCL7/MCP‐3, and CCL20/MIP3α protein levels increased during virus‐positive sick weeks. Only a subset of cytokines (IFN‐γ, CXCL8, CCL2, CCL4, CCL5, and CCL20) correlated with self‐reported respiratory tract symptoms. While many aspirates were dilute and showed no mRNA signal, viral infection significantly increased the number of samples that were positive for IFN‐λ1, IFN‐λ2/3, TLR3, RIG‐I, and IRF7 mRNA. Conclusions and clinical relevance We conclude that in children with asthma, naturally occurring viral infections apparently induce a robust innate immune response including expression of specific chemokines, IFNs, and IFN‐responsive genes.
Collapse
Affiliation(s)
- T C Lewis
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Schneider D, Hong JY, Bowman ER, Chung Y, Nagarkar DR, McHenry CL, Goldsmith AM, Bentley JK, Lewis TC, Hershenson MB. Macrophage/epithelial cell CCL2 contributes to rhinovirus-induced hyperresponsiveness and inflammation in a mouse model of allergic airways disease. Am J Physiol Lung Cell Mol Physiol 2012. [PMID: 23204071 DOI: 10.1152/ajplung.00182.2012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Human rhinovirus (HRV) infections lead to exacerbations of lower airways disease in asthmatic patients but not in healthy individuals. However, underlying mechanisms remain to be completely elucidated. We hypothesized that the Th2-driven allergic environment enhances HRV-induced CC chemokine production, leading to asthma exacerbations. Ovalbumin (OVA)-sensitized and -challenged mice inoculated with HRV showed significant increases in the expression of lung CC chemokine ligand (CCL)-2/monocyte chemotactic protein (MCP)-1, CCL4/macrophage inflammatory protein (MIP)-1β, CCL7/MCP-3, CCL19/MIP-3β, and CCL20/MIP3α compared with mice treated with OVA alone. Inhibition of CCL2 with neutralizing antibody significantly attenuated HRV-induced airways inflammation and hyperresponsiveness in OVA-treated mice. Immunohistochemical stains showed colocalization of CCL2 with HRV in epithelial cells and CD68-positive macrophages, and flow cytometry showed increased CCL2(+), CD11b(+) cells in the lungs of OVA-treated, HRV-infected mice. Compared with lung macrophages from naïve mice, macrophages from OVA-exposed mice expressed significantly more CCL2 in response to HRV infection ex vivo. Pretreatment of mouse lung macrophages and BEAS-2B human bronchial epithelial cells with interleukin (IL)-4 and IL-13 increased HRV-induced CCL2 expression, and mouse lung macrophages from IL-4 receptor knockout mice showed reduced CCL2 expression in response to HRV, suggesting that exposure to these Th2 cytokines plays a role in the altered HRV response. Finally, bronchoalveolar macrophages from children with asthma elaborated more CCL2 upon ex vivo exposure to HRV than cells from nonasthmatic patients. We conclude that CCL2 production by epithelial cells and macrophages contributes to HRV-induced airway hyperresponsiveness and inflammation in a mouse model of allergic airways disease and may play a role in HRV-induced asthma exacerbations.
Collapse
Affiliation(s)
- Dina Schneider
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Teran LM, Rüggeberg S, Santiago J, Fuentes-Arenas F, Hernández JL, Montes-Vizuet AR, Xinping L, Franz T. Immune Response to Seasonal Influenza A Virus Infection: A Proteomic Approach. Arch Med Res 2012; 43:464-9. [DOI: 10.1016/j.arcmed.2012.08.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 08/06/2012] [Indexed: 10/27/2022]
|
21
|
Gotera J, Giuffrida M, Mavarez A, Pons H, Bermudez J, Maldonado M, Espina LM, Mosquera J, Valero N. Respiratory syncytial virus infection increases regulated on activation normal T cell expressed and secreted and monocyte chemotactic protein 1 levels in serum of patients with asthma and in human monocyte cultures. Ann Allergy Asthma Immunol 2012; 108:316-20. [PMID: 22541401 DOI: 10.1016/j.anai.2012.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 02/28/2012] [Accepted: 03/05/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection is associated to episodic exacerbations of asthma involving alveolar macrophages and chemokine production. OBJECTIVE The aim of this study was to determine the circulating levels of monocyte chemotactic protein 1 (MCP-1), regulated on activation normal T cell expressed and secreted (RANTES), and substance P (SP) in patients with and without asthma with acute respiratory RSV infection and the chemokine profile in RSV- infected monocyte cultures from normal individuals and individuals with asthma. METHODS In this regard, 31 adult patients with acute respiratory infection (15 patients with asthma) were studied. MCP-1, RANTES and SP were measured in serum and in supernatants from monocyte cultures by enzyme-linked immunosorbent assay (ELISA). RESULTS Increased levels of MCP-1 and RANTES were observed in serum from patients with asthma related to RSV infection. RSV-infected monocyte cultures from healthy individuals showed increased content of those chemokines, and monocyte cultures from patients with asthma showed increased expression of MCP-1. CONCLUSION These data show that RSV infection induces increased circulating level of chemokines in patients with asthma, and this finding could be mediated in part by the interaction virus-monocyte.
Collapse
Affiliation(s)
- Jenifer Gotera
- Instituto de Investigaciones Clínicas Dr. Américo Negrette, Facultad de Medicina, Universidad del Zulia, Maracaibo, Venezuela
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Host regulatory network response to infection with highly pathogenic H5N1 avian influenza virus. J Virol 2011; 85:10955-67. [PMID: 21865398 DOI: 10.1128/jvi.05792-11] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
During the last decade, more than half of humans infected with highly pathogenic avian influenza (HPAI) H5N1 viruses have died, yet virus-induced host signaling has yet to be clearly elucidated. Airway epithelia are known to produce inflammatory mediators that contribute to HPAI H5N1-mediated pathogenicity, but a comprehensive analysis of the host response in this cell type is lacking. Here, we leveraged a system approach to identify and statistically validate signaling subnetworks that define the dynamic transcriptional response of human bronchial epithelial cells after infection with influenza A/Vietnam/1203/2004 (H5N1, VN1203). Importantly, we validated a subset of transcripts from one subnetwork in both Calu-3 cells and mice. A more detailed examination of two subnetworks involved in the immune response and keratinization processes revealed potential novel mediators of HPAI H5N1 pathogenesis and host response signaling. Finally, we show how these results compare to those for a less virulent strain of influenza virus. Using emergent network properties, we provide fresh insight into the host response to HPAI H5N1 virus infection and identify novel avenues for perturbation studies and potential therapeutic interventions for fatal HPAI H5N1 disease.
Collapse
|
23
|
Current world literature. Curr Opin Allergy Clin Immunol 2010; 10:161-6. [PMID: 20357579 DOI: 10.1097/aci.0b013e32833846d5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|