1
|
Pereira De Oliveira R, Droillard C, Devouassoux G, Rosa-Calatrava M. In vitro models to study viral-induced asthma exacerbation: a short review for a key issue. FRONTIERS IN ALLERGY 2025; 6:1530122. [PMID: 40224321 PMCID: PMC11987631 DOI: 10.3389/falgy.2025.1530122] [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: 11/18/2024] [Accepted: 02/27/2025] [Indexed: 04/15/2025] Open
Abstract
Asthma is a heterogenous inflammatory bronchial disease involving complex mechanisms, several inflammatory pathways, and multiples cell-type networks. Bronchial inflammation associated to asthma is consecutive to multiple aggressions on epithelium, such as microbiologic, pollutant, and antigenic agents, which are responsible for both T2 and non-T2 inflammatory responses and further airway remodeling. Because asthma physiopathology involves multiple crosstalk between several cell types from different origins (epithelial, mesenchymal, and immune cells) and numerous cellular effectors, no single and/or representative in vitro model is suitable to study the overall of this disease. In this short review, we present and discuss the advantages and limitations of different in vitro models to decipher different aspects of virus-related asthma physiopathology and exacerbation.
Collapse
Affiliation(s)
- Rémi Pereira De Oliveira
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec- Université Laval, Québec, QC, Canada
- International Research Laboratory RESPIVIR France – Canada, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Clément Droillard
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec- Université Laval, Québec, QC, Canada
- International Research Laboratory RESPIVIR France – Canada, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Gilles Devouassoux
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec- Université Laval, Québec, QC, Canada
- International Research Laboratory RESPIVIR France – Canada, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Department of Respiratory Diseases, CIERA, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon et CRISALIS/F-CRIN INSERM Network, Lyon, France
| | - Manuel Rosa-Calatrava
- CIRI, Centre International de Recherche en Infectiologie, Team VirPath, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- International Research Laboratory RESPIVIR France - Canada, Centre Hospitalier Universitaire de Québec- Université Laval, Québec, QC, Canada
- International Research Laboratory RESPIVIR France – Canada, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Virnext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- Centre de Recherche en Infectiologie du Centre Hospitalier Universitaire de Québec - Université Laval, Faculté de Médecine, Département de Pédiatrie de l’Université Laval, Québec, QC, Canada
| |
Collapse
|
2
|
Li Q, Shan X, Yuan Y, Ye W, Fang X. Shegan-Mahuang decoction ameliorates cold-induced asthma via regulating the proliferation and apoptosis of airway smooth muscle cells through TAS2R10: An in vivo and in vitro study. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118504. [PMID: 38950796 DOI: 10.1016/j.jep.2024.118504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shegan-Mahuang Decoction (SMD) is a classical formula that has been used to effectively treat cold-induced asthma (CA) for 1800 years. Airway smooth muscle cells (ASMCs) play a crucial role in airway remodeling of CA and can be modulated through bitter taste-sensing type 2 receptors (TAS2Rs). Given that SMD contains numerous bitter herbs and TAS2R10 expression in ASMCs remains consistently high, it is pertinent to explore whether SMD regulates ASMCs via TAS2R10 to exert its CA mechanism. AIM OF THE STUDY This study investigated the efficacy as well as the potential mechanism of SMD in CA. MATERIALS AND METHODS In this study, experiments in vivo were conducted using the CA rat model induced by ovalbumin (OVA) along with cold stimulation. The effects of SMD and TAS2R10 expression in CA rats were evaluated using the following methods: clinical symptoms, weights, pathological staining, immunofluorescence staining (IF), enzyme-linked immunosorbent assay (ELISA), real-time quantitative polymerase chain reaction (RT-qPCR) and Western blot (WB). Assays in vitro including cell counting Kit-8 (CCK-8), ELISA, flow cytometry, TUNEL staining, RT-qPCR and WB were performed to investigate potential mechanism of SMD on the proliferation and apoptosis of ASMCs through upregulation of TAS2R10. RESULTS The administration of SMD resulted in a notable improvement in the symptoms, trends in weight, airway inflammation and airway remodeling observed in CA rats with upregulated TAS2R10. Mechanistically, we furtherly confirmed that SMD inhibits p70S6K/CyclinD1 pathway by upregulating TAS2R10. SMD furthermore blocked the G0/G1 phase, suppressed the proliferation and inducted apoptosis in ASMCs induced by platelet-derived growth factor-BB (PDGF-BB). Erythromycin (EM), a TAS2R10 agonist, can intensify these effects. CONCLUSIONS SMD significantly ameliorates CA by upregulating TAS2R10 and inhibiting the p70S6K/CyclinD1 pathway, thereby modulating ASMCs' proliferation and apoptosis. Inspired by the Five Flavors Theory of Traditional Chinese Medicine, this study provides an updated treatment perspective for treating CA.
Collapse
Affiliation(s)
- Qiuhui Li
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Xiaoxiao Shan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Yamei Yuan
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Weidong Ye
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Xiangming Fang
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
3
|
Xue J, Liu Z, Xie B, Dong R, Wu J, Wu Y, Xu Z, Tian Y, Wei Y, Geng Z, Lu L, Liu Y, Xie J, Yang P. Probiotic nucleotides increase IL-10 expression in airway macrophages to mitigate airway allergy. Inflamm Res 2024; 73:1919-1930. [PMID: 39235607 DOI: 10.1007/s00011-024-01940-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/16/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Dysfunctional immune regulation plays a crucial role in the pathogenesis of airway allergies. Macrophages are one of the components of the immune regulation cells. The aim of this study is to elucidate the role of lysine demethylase 5 A (KDM5A) in maintaining macrophages' immune regulatory ability. METHODS DNA was extracted from Lactobacillus rhamnosus GG to be designated as LgDNA. LgDNA was administered to the mice through nasal instillations. M2 macrophages (M2 cells) were isolated from the airway tissues using flow cytometry. RESULTS We found that airway M2 cells of mice with airway Th2 polarization had reduced amounts of IL-10 and KDM5A. Mice with Kdm5a deficiency in M2 cells showed the airway Th2 polarization. The expression of Kdm5a in airway M2 cells was enhanced by nasal instillations containing LgDNA. KDM5A mediated the effects of LgDNA on inducing the Il10 expression in airway M2 cells. Administration of LgDNA mitigated experimental airway allergy. CONCLUSIONS M2 macrophages in the airway tissues of mice with airway allergy show low levels of KDM5A. By upregulating KDM5A expression, LgDNA can increase Il10 expression and reconcile airway Th2 polarization.
Collapse
Affiliation(s)
- Jinmei Xue
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China
| | - Bailing Xie
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy and Immunology, Shenzhen University School of Medicine, Room A7-509 at Lihu Campus. 1066 Xueyuan Blvd., Shenzhen, China
| | - Rui Dong
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Juan Wu
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Yisha Wu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Zhihan Xu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Yuhe Tian
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Yao Wei
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Zhigang Geng
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Lei Lu
- Department of Otolaryngology, Head and Neck Surgery, Second Hospital, Shanxi Medical University, Taiyuan, China
- Shanxi Key Laboratory of Rapid Diagnosis and Precision Treatment of Airway Allergic Diseases, Taiyuan, China
| | - Yu Liu
- Department of General Practice Medicine, Third Hospital of Shenzhen University, Shenzhen, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Shanxi Medical University, Taiyuan, 030001, China.
| | - Pingchang Yang
- State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University and Institute of Allergy and Immunology, Shenzhen University School of Medicine, Room A7-509 at Lihu Campus. 1066 Xueyuan Blvd., Shenzhen, China.
| |
Collapse
|
4
|
Rajaiah R, Pandey K, Acharya A, Ambikan A, Kumar N, Guda R, Avedissian SN, Montaner LJ, Cohen SM, Neogi U, Byrareddy SN. Differential immunometabolic responses to Delta and Omicron SARS-CoV-2 variants in golden syrian hamsters. iScience 2024; 27:110501. [PMID: 39171289 PMCID: PMC11338146 DOI: 10.1016/j.isci.2024.110501] [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: 08/24/2023] [Revised: 02/07/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
Delta (B.1.617.2) and Omicron (B.1.1.529) variants of SARS-CoV-2 represents unique clinical characteristics. However, their role in altering immunometabolic regulations during acute infection remains convoluted. Here, we evaluated the differential immunopathogenesis of Delta vs. Omicron variants in Golden Syrian hamsters (GSH). The Delta variant resulted in higher virus titers in throat swabs and the lungs and exhibited higher lung damage with immune cell infiltration than the Omicron variant. The gene expression levels of immune mediators and metabolic enzymes, Arg-1 and IDO1 in the Delta-infected lungs were significantly higher compared to Omicron. Further, Delta/Omicron infection perturbed carbohydrates, amino acids, nucleotides, and TCA cycle metabolites and was differentially regulated compared to uninfected lungs. Collectively, our data provide a novel insight into immunometabolic/pathogenic outcomes for Delta vs. Omicron infection in the GSH displaying concordance with COVID-19 patients associated with inflammation and tissue injury during acute infection that offered possible new targets to develop potential therapeutics.
Collapse
Affiliation(s)
- Rajesh Rajaiah
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kabita Pandey
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anoop Ambikan
- The Systems Virology Lab, Department of Laboratory Medicine, Division of Clinical Microbiology, ANA Futura, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Narendra Kumar
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Reema Guda
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sean N. Avedissian
- Antiviral Pharmacology Laboratory, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Luis J. Montaner
- Vaccine and Immunotherapy Center, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Samuel M. Cohen
- Havlik Wall Professor of Oncology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ujjwal Neogi
- The Systems Virology Lab, Department of Laboratory Medicine, Division of Clinical Microbiology, ANA Futura, Karolinska Institutet, 141 52 Stockholm, Sweden
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
- Havlik Wall Professor of Oncology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
5
|
Chen G, Zheng Y, Wu N, Yang X, Qu S. Human beta defensin 3 knockdown inhibits the proliferation and migration of airway smooth muscle cells through regulating the PI3K/AKT signaling pathway. Mol Immunol 2024; 168:38-46. [PMID: 38422885 DOI: 10.1016/j.molimm.2024.02.013] [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: 11/01/2023] [Revised: 02/04/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
Asthma, a common pediatric pulmonary disease, significantly affects children's healthy development. This study aimed to investigate the functions of human β defensin-3 (HBD-3) in asthma progression. For this purpose, blood samples from asthmatic and healthy children were collected. Moreover, the airway smooth muscle cells (ASMCs) were treated with platelet-derived growth factor BB (PDGF-BB) to develop an in vitro asthma model, then evaluated cell viability and migration via CCK-8 and transwell assays. The mRNA levels of interferon γ (INF-γ), interleukin 4 (IL-4), interleukin 10 (IL-10), alpha-smooth muscle actin (α-SMA), HBD-3, and the protein levels of phosphatidylinositol 3-kinase (PI3K) along with protein kinase B (AKT) were detected. Similarly, the N6-methyladenosine (m6A) content in the ASMCs and m6A levels of HBD-3 were also measured. Results indicated an upregulated HBD-3 in the asthmatic children. The ASMCs were found to be stimulated by PDGF-BB, in addition to the promotion of cell viability and migration. The INF-γ, IL-4, and α-SMA levels were reduced, while IL-10 was elevated in PDGF-BB-stimulated ASMCs. Silencing HBD-3 in PDGF-BB stimulated ASMCs was found to exert the opposite effect by inhibiting cell viability and migration, enhancing the levels of INF-γ, IL-4, and α-SMA, while the IL-10 levels were found to decline. PDGF-BB stimulation of ASMCs resulted in activation of the PI3K/AKT signaling pathway, which was blocked post HBD-3 silencing, while the role of si-hBD in PDGF-BB stimulated ASMCs was neutralized post-treatment with IGF-1. Finally, it was found that METTL3 overexpression prominently upregulated the m6A levels of HBD-3 and decreased the mRNA expression and stability of HBD-3 in the PDGF-BB-stimulated ASMCs. The study concluded that METTL3-mediated HBD-3 participates in the progression of asthma through the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Guiying Chen
- Department of Pediatrics, Second Affiliated Hospital of Harbin Medical University, China; Department of Emergency, Sixth Affiliated Hospital of Harbin Medical University(Jiangnan Courtyard), China
| | - Yuling Zheng
- Department of Pediatrics, Second Affiliated Hospital of Harbin Medical University, China
| | - Nan Wu
- Department of Emergency, Sixth Affiliated Hospital of Harbin Medical University(Jiangnan Courtyard), China
| | - Xia Yang
- Department of Respiratory, Sixth Affiliated Hospital of Harbin Medical University (Jiangnan Courtyard), China
| | - Shuqiang Qu
- Department of Pediatrics, Second Affiliated Hospital of Harbin Medical University, China.
| |
Collapse
|
6
|
Zhou Z, Zhang H, Yuan Y. Association between IL10 rs1800896 polymorphism and risk of pediatric asthma: A meta-analysis. THE CLINICAL RESPIRATORY JOURNAL 2023; 17:1276-1285. [PMID: 37937689 PMCID: PMC10730469 DOI: 10.1111/crj.13714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/28/2023] [Accepted: 10/14/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION Asthma is a common chronic condition in children. Several studies have explored the potential association between IL10 rs1800896 polymorphism and the risk of asthma in children, but the findings have been inconsistent. To address these discrepancies, we conducted a systematic review and meta-analysis to assess the relationship between IL10 rs1800896 polymorphisms and the susceptibility to pediatric asthma. METHODS A literature search was conducted in PubMed, Scopus, Web of Science, and CNKI databases to identify eligible studies through April 2022. Meta-analysis was then performed using five genetic models: dominant, recessive, homozygous, heterozygous, and allele. RESULTS A total of 12 studies comprising 1645 cases along with 1447 controls were included in this meta-analysis. It was found that rs1800896 was not associated significantly with susceptibility to childhood asthma in all genetic models investigated. Subgroup analysis based on the ethnic background of the subjects revealed that rs1800896 was significantly linked to a lower risk of pediatric asthma among Asians in the homozygous model (OR = 0.311, 95% CI = 0.152-0.637, P = 0.001) and in the recessive model (OR = 0.585, 95% CI = 0.405-0.846, P = 0.004), whereas no significant relationship was observed in Egyptians (P > 0.05). CONCLUSION In conclusion, IL10 rs1800896 polymorphism may be useful as a predictive marker for childhood asthma in Asians, although further studies are needed to validate the study results.
Collapse
Affiliation(s)
- Zhihong Zhou
- School of NursingHebi PolytechnicHebiChina
- SeHan UniversityYeongam‐gunJeollanam‐doRepublic of Korea
| | - Hui Zhang
- Department of HepatopathyHunan Children's HospitalChangshaChina
| | - Yuanhong Yuan
- Emergency CenterHunan Children's HospitalChangshaChina
| |
Collapse
|
7
|
Shimoda M, Inagaki T, Davis RR, Merleev A, Tepper CG, Maverakis E, Izumiya Y. Virally encoded interleukin-6 facilitates KSHV replication in monocytes and induction of dysfunctional macrophages. PLoS Pathog 2023; 19:e1011703. [PMID: 37883374 PMCID: PMC10602306 DOI: 10.1371/journal.ppat.1011703] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic double-stranded DNA virus and the etiologic agent of Kaposi's sarcoma and hyperinflammatory lymphoproliferative disorders. Understanding the mechanism by which KSHV increases the infected cell population is crucial for curing KSHV-associated diseases. Using scRNA-seq, we demonstrate that KSHV preferentially infects CD14+ monocytes, sustains viral lytic replication through the viral interleukin-6 (vIL-6), which activates STAT1 and 3, and induces an inflammatory gene expression program. To study the role of vIL-6 in monocytes upon KSHV infection, we generated recombinant KSHV with premature stop codon (vIL-6(-)) and its revertant viruses (vIL-6(+)). Infection of the recombinant viruses shows that both vIL-6(+) and vIL-6(-) KSHV infection induced indistinguishable host anti-viral response with STAT1 and 3 activations in monocytes; however, vIL-6(+), but not vIL-6(-), KSHV infection promoted the proliferation and differentiation of KSHV-infected monocytes into macrophages. The macrophages derived from vIL-6(+) KSHV infection showed a distinct transcriptional profile of elevated IFN-pathway activation with immune suppression and were compromised in T-cell stimulation function compared to those from vIL-6(-) KSHV infection or uninfected control. Notably, a viral nuclear long noncoding RNA (PAN RNA), which is required for sustaining KSHV gene expression, was substantially reduced in infected primary monocytes upon vIL-6(-) KSHV infection. These results highlight the critical role of vIL-6 in sustaining KSHV transcription in primary monocytes. Our findings also imply a clever strategy in which KSHV utilizes vIL-6 to secure its viral pool by expanding infected monocytes via differentiating into longer-lived dysfunctional macrophages. This mechanism may facilitate KSHV to escape from host immune surveillance and to support a lifelong infection.
Collapse
Affiliation(s)
- Michiko Shimoda
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
- UC Davis Comprehensive Cancer Center, Sacramento, California, United States of America
| | - Tomoki Inagaki
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
| | - Ryan R. Davis
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, California, United States of America
| | - Alexander Merleev
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
| | - Clifford G. Tepper
- UC Davis Comprehensive Cancer Center, Sacramento, California, United States of America
- Department of Biochemistry and Molecular Medicine, School of Medicine, UC Davis, Sacramento, California, United States of America
| | - Emanual Maverakis
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
- UC Davis Comprehensive Cancer Center, Sacramento, California, United States of America
| | - Yoshihiro Izumiya
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, California, United States of America
- UC Davis Comprehensive Cancer Center, Sacramento, California, United States of America
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, United States of America
| |
Collapse
|
8
|
Rolland-Debord C, Piéroni L, Bejar F, Milon A, Choinier P, Blin E, Bravais J, Halitim P, Letellier A, Camuset J, Parrot A, Fajac A, Cadranel J. Cell and cytokine analyses from bronchoalveolar lavage in non-critical COVID-19 pneumonia. Intern Emerg Med 2023; 18:1723-1732. [PMID: 37353659 DOI: 10.1007/s11739-023-03341-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/05/2023] [Indexed: 06/25/2023]
Abstract
Cell and cytokine analyses from bronchoalveolar lavage (BAL) in non-critically ill patients with COVID-19 pneumonia are poorly described. This study focused on patients hospitalized in the non-intensive care unit for either suspected COVID-19 pneumonia or persistent respiratory symptoms following proven COVID-19 pneumonia. Overall, 54 patients who underwent BAL between April 2020 and February 2021 for suspected or follow-up of proven COVID-19 pneumonia were included. Based on SARS-CoV-2 polymerase chain reaction test results and clinical follow-up, three pulmonary disease groups were defined: non-COVID-19 (n = 20), acute COVID-19 (n = 13), and post-COVID-19 (n = 24) pneumonia patients. Cytological and cytokine analyses were performed on BAL fluid (IL-1β, IL-6, IL-8, IL-10, TNF-α, IFN-γ, HGF, and TGF-β), with investigators blinded to the patient groups. Lymphocytic alveolitis with plasmocytes was observed in acute COVID-19 pneumonia, returning to normal post-COVID-19. The highest cytokine levels were observed in COVID-19 patients, with significantly increased IFN-γ, IL-10, and HGF levels compared to non-COVID-19 patients, while significantly decreased IL-6, IL-8, IL-10, IFN-γ, TNF-α, and HGF levels were noted in post-COVID-19 patients. In COVID-19 patients, correlations between IL-10, TNF-α and IFN-γ concentrations were found. Lymphocytic alveolitis with plasmacytosis was found in non-critical COVID-19 pneumonia This alveolitis is associated with the presence of IL-6, IL-8, IL-10, TNF-α, IFN-γ and HGF. Alveolitis and cytokines levels decreased in post-COVID-19 pneumonia.
Collapse
Affiliation(s)
- Camille Rolland-Debord
- Department of Pneumology and Thoracic Oncology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France.
- Department of Pneumology, Service de Pneumologie, CHU Gabriel Montpied, Université Clermont Auvergne, 53 rue Montalembert, 63000, Clermont-Ferrand, France.
| | - Laurence Piéroni
- Department of Biochemistry, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Farah Bejar
- Department of Biochemistry, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Audrey Milon
- Department of Radiology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Pascaline Choinier
- Department of Pneumology and Thoracic Oncology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Emmanuelle Blin
- Department of Pneumology and Thoracic Oncology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Juliette Bravais
- Department of Pneumology and Thoracic Oncology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Pierre Halitim
- Department of Pneumology and Thoracic Oncology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Alice Letellier
- Department of Pneumology and Thoracic Oncology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Juliette Camuset
- Department of Thoracic and Vascular Surgery, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Antoine Parrot
- Department of Pneumology and Thoracic Oncology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Anne Fajac
- Department of Pathology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| | - Jacques Cadranel
- Department of Pneumology and Thoracic Oncology, AP-HP Hôpital Tenon, Sorbonne Université, Paris, France
| |
Collapse
|
9
|
Xia Y, Yang Q, Wu SY, Wu Z, Li Q, Du J. Interferon lambda modulates proinflammatory cytokines production in PBMCs from patients with chronic kidney disease. Hum Immunol 2023; 84:464-470. [PMID: 37394297 DOI: 10.1016/j.humimm.2023.06.001] [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: 12/27/2022] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 07/04/2023]
Abstract
BACKGROUND CKD is a major cause of morbidity and mortality worldwide. Considerable evidence now indicates that renal inflammation plays a central role in the initiation and progression of CKD. Recent investigations have demonstrated that IFNλ plays an important role in the pathogenesis of autoimmune and inflammatory diseases. However, the association of IFNλ with CKD is still poorly understood. OBJECTIVE To analyze the correlation between IFNλ levels and pro-inflammatory cytokines, and to investigate the effect of IFNλ on PBMCs in patients with CKD. METHODS PBMCs were harvested from patients with CKD and healthy controls for measuring the expression level of inflammatory cytokines by RT-qPCR. Spearman correlation test was used to analyze correlation between IFNλ and cytokines as well as eGFR. PBMCs from healthy individuals and CKD patients were subjected to IFNλ protein stimulation. IL6, TNFα, IL10, ISG15 and MX1 mRNA level were measured by RT-PCR, STAT1 and phosphorylated STAT1 protein level were measured by Western blot. RESULTS Patients with CKD showed higher levels of IFNλ in PBMCs compared to healthy controls. IFNλ mRNA levels were associated with cytokines and eGFR. The transcription of IL6, TNFα, and IL10 was significantly increased in healthy human PBMCs after IFNλ stimulation. In addition, IFNλ acts on PBMCs by p-STAT1 and ISG15 as well as MX1. CONCLUSION High expression of IFNλ was found in CKD patients and was associated with eGFR and disease-related cytokines. More importantly, IFNλ promoted the expression of pro-inflammatory cytokines in PBMCs, suggesting a potential pro-inflammatory role of IFNλ in CKD.
Collapse
Affiliation(s)
- Yuhao Xia
- Weifang Medical University, Shandong, China; Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Qiannan Yang
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Shang Ying Wu
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Zhicheng Wu
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Qian Li
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| | - Jing Du
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
10
|
Shimoda M, Inagaki T, Davis RR, Merleev A, Tepper CG, Maverakis E, Izumiya Y. KSHV uses viral IL6 to expand infected immunosuppressive macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.05.531224. [PMID: 36945595 PMCID: PMC10028810 DOI: 10.1101/2023.03.05.531224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is an oncogenic double-stranded DNA virus and the etiologic agent of Kaposi's sarcoma and hyperinflammatory lymphoproliferative disorders. Understanding the mechanism by which KSHV increases the infected cell population is crucial for curing KSHV-associated diseases. Here we demonstrate that KSHV preferentially infects CD14 + monocytes and sustains viral replication through the viral interleukin-6 (vIL6)-mediated activation of STAT1 and 3. Using vIL6-sufficient and vIL6-deficient recombinant KSHV, we demonstrated that vIL6 plays a critical role in promoting the proliferation and differentiation of KSHV-infected monocytes into macrophages. The macrophages derived from vIL6-sufficient KSHV infection showed a distinct transcriptional profile of elevated IFN-pathway activation with immune suppression and were compromised in T-cell stimulation function compared to those from vIL6-deficient KSHV infection or uninfected control. These results highlight a clever strategy, in which KSHV utilizes vIL6 to secure its viral pool by expanding infected dysfunctional macrophages. This mechanism also facilitates KSHV to escape from host immune surveillance and to establish a lifelong infection. 160. Summary KSHV causes multiple inflammatory diseases, however, the underlying mechanism is not clear. Shimoda et al. demonstrate that KSHV preferentially infects monocytes and utilizes virally encoded interleukin-6 to expand and deregulate infected monocytes. This helps the virus escape from host immune surveillance.
Collapse
|
11
|
Liang W, Qi Y, Yi H, Mao C, Meng Q, Wang H, Zheng C. The Roles of Adipose Tissue Macrophages in Human Disease. Front Immunol 2022; 13:908749. [PMID: 35757707 PMCID: PMC9222901 DOI: 10.3389/fimmu.2022.908749] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/12/2022] [Indexed: 01/02/2023] Open
Abstract
Macrophages are a population of immune cells functioning in antigen presentation and inflammatory response. Research has demonstrated that macrophages belong to a cell lineage with strong plasticity and heterogeneity and can be polarized into different phenotypes under different microenvironments or stimuli. Many macrophages can be recruited by various cytokines secreted by adipose tissue. The recruited macrophages further secrete various inflammatory factors to act on adipocytes, and the interaction between the two leads to chronic inflammation. Previous studies have indicated that adipose tissue macrophages (ATMs) are closely related to metabolic diseases like obesity and diabetes. Here, we will not only conclude the current progress of factors affecting the polarization of adipose tissue macrophages but also elucidate the relationship between ATMs and human diseases. Furthermore, we will highlight its potential in preventing and treating metabolic diseases as immunotherapy targets.
Collapse
Affiliation(s)
- Weizheng Liang
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China.,Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yanxu Qi
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hongyang Yi
- National Clinical Research Centre for Infectious Diseases, The Third People's Hospital of Shenzhen and The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Chenyu Mao
- School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Qingxue Meng
- Central Laboratory, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Hao Wang
- Shenzhen Key Laboratory, Shenzhen University General Hospital, Shenzhen, China.,Department of Obstetrics and Gynecology, Shenzhen University General Hospital, Shenzhen, China
| | - Chunfu Zheng
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
12
|
LncTRPM2-AS inhibits TRIM21-mediated TRPM2 ubiquitination and prevents autophagy-induced apoptosis of macrophages in asthma. Cell Death Dis 2021; 12:1153. [PMID: 34903714 PMCID: PMC8668916 DOI: 10.1038/s41419-021-04437-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/09/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022]
Abstract
Long non-coding RNAs (lncRNAs) play a crucial role in macrophage development but little is known about their role in asthma. Here, we investigated the role of lncRNA lncTRPM2-AS in asthma and found that lncTRPM2-AS participates in the promotion of macrophage inflammation. Downregulation of lncTRPM2-AS promoted apoptosis and inhibited proliferation and production of cytokines including IL-1β, IL-4, IL-6, IL-10, TNF-α, and TGF-β. RNA-immunoprecipitation and mass spectrometry indicated that the protein TRPM2 interacted with both lncTRPM2-AS and the E3 ubiquitin ligase TRIM21. LncTRPM2-AS silencing enhanced the interaction between TRIM21 and TRPM2, resulting in elevated levels of ubiquitin-related degradation of TRPM2. Mutation analysis indicated that TRPM2 K1218 is a key site for TRIM21-dependent ubiquitination. Downregulation of lncTRPM2-AS significantly decreased intracellular calcium levels by restraining TRPM2 protein expression, which in turn decreased ROS levels and increased autophagy to promote macrophage apoptosis and reduce cytokine production, together inhibiting macrophage inflammation. Taken together, our findings demonstrate that lncTRPM2-AS blocks the ubiquitination of TRPM2 via TRIM21 and inhibits autophagy-induced apoptosis which may contribute to macrophage inflammation in asthma.
Collapse
|
13
|
Network Pharmacology-Based Analysis of the Underlying Mechanism of Hyssopus cuspidatus Boriss. for Antiasthma: A Characteristic Medicinal Material in Xinjiang. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:7671247. [PMID: 34880921 PMCID: PMC8648465 DOI: 10.1155/2021/7671247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 10/26/2021] [Indexed: 12/24/2022]
Abstract
Background Hyssopus cuspidatus Boriss. (Shen Xiang Cao (SXC)), a traditional medicine herb in Xinjiang, has a long history of being used by minorities to treat asthma. However, its active antiasthmatic compounds and underlying mechanism of action are still unknown. The aim of this study was to investigate the bioactive compounds and explore the molecular mechanism of SCX in the treatment of asthma using network pharmacology. Methods The compounds of SCX were collected by a literature search, and Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and SwissTargetPrediction were used to predict targets and screen active compounds. Moreover, asthma-related targets were obtained based on DisGeNET, Herb, and GeneCards databases, and a protein-protein interaction (PPI) network was built by the STRING database. Furthermore, the topological analysis of the PPI and SXC-compound-target networks were analyzed and established by Cytoscape software. Finally, the RStudio software package was used for carrying out Gene Ontology (GO) function enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. AutoDock tools and AutoDock Vina were used to molecularly dock the active compounds and key targets. Results A total of 8 active compounds and 258 potential targets related to SXC were predicted, and PPI network screened out key targets, including IL-6, JUN, TNF, IL10, and CXCL8. GO enrichment analysis involved cell responses to reactive oxygen species, oxidative stress, chemical stress, etc. In addition, KEGG pathway analysis showed that SXC effectively treated asthma through regulation of mitogen-activated protein kinases (MAPK) signaling pathways, interleukin 17 (IL-17) signaling pathways, toll-like receptor (TLR) signaling pathways, and tumor necrosis factor (TNF) signaling pathways. Conclusion The preliminary study that was based on multiple compounds, multiple targets, and multiple pathways provides a scientific basis for further elucidating the molecules involved and the underlying antiasthma-related mechanisms of SXC.
Collapse
|
14
|
Pathak GA, Singh K, Miller-Fleming TW, Wendt FR, Ehsan N, Hou K, Johnson R, Lu Z, Gopalan S, Yengo L, Mohammadi P, Pasaniuc B, Polimanti R, Davis LK, Mancuso N. Integrative genomic analyses identify susceptibility genes underlying COVID-19 hospitalization. Nat Commun 2021; 12:4569. [PMID: 34315903 PMCID: PMC8316582 DOI: 10.1038/s41467-021-24824-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 07/07/2021] [Indexed: 12/11/2022] Open
Abstract
Despite rapid progress in characterizing the role of host genetics in SARS-Cov-2 infection, there is limited understanding of genes and pathways that contribute to COVID-19. Here, we integrate a genome-wide association study of COVID-19 hospitalization (7,885 cases and 961,804 controls from COVID-19 Host Genetics Initiative) with mRNA expression, splicing, and protein levels (n = 18,502). We identify 27 genes related to inflammation and coagulation pathways whose genetically predicted expression was associated with COVID-19 hospitalization. We functionally characterize the 27 genes using phenome- and laboratory-wide association scans in Vanderbilt Biobank (n = 85,460) and identified coagulation-related clinical symptoms, immunologic, and blood-cell-related biomarkers. We replicate these findings across trans-ethnic studies and observed consistent effects in individuals of diverse ancestral backgrounds in Vanderbilt Biobank, pan-UK Biobank, and Biobank Japan. Our study highlights and reconfirms putative causal genes impacting COVID-19 severity and symptomology through the host inflammatory response.
Collapse
Affiliation(s)
- Gita A Pathak
- Yale School of Medicine, Department of Psychiatry, Division of Human Genetics, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Kritika Singh
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tyne W Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Frank R Wendt
- Yale School of Medicine, Department of Psychiatry, Division of Human Genetics, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Nava Ehsan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Kangcheng Hou
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Ruth Johnson
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, USA
| | - Zeyun Lu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shyamalika Gopalan
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Loic Yengo
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Pejman Mohammadi
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
- Scripps Translational Science Institute, The Scripps Research Institute, La Jolla, CA, USA
| | - Bogdan Pasaniuc
- Departments of Computational Medicine, Human Genetics, Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Renato Polimanti
- Yale School of Medicine, Department of Psychiatry, Division of Human Genetics, New Haven, CT, USA
- Veteran Affairs Connecticut Healthcare System, West Haven, CT, USA
| | - Lea K Davis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicholas Mancuso
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Turan N, van der Veen TA, Draijer C, Fattahi F, ten Hacken NH, Timens W, van Oosterhout AJ, van den Berge M, Melgert BN. Neutrophilic Asthma Is Associated With Smoking, High Numbers of IRF5+, and Low Numbers of IL10+ Macrophages. FRONTIERS IN ALLERGY 2021; 2:676930. [PMID: 35387061 PMCID: PMC8974785 DOI: 10.3389/falgy.2021.676930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/26/2021] [Indexed: 11/13/2022] Open
Abstract
Asthma is a heterogenous disease with different inflammatory subgroups that differ in disease severity. This disease variation is hampering treatment and development of new treatment strategies. Macrophages may contribute to asthma phenotypes by their ability to activate in different ways, i.e., T helper cell 1 (Th1)-associated, Th2-associated, or anti-inflammatory activation. It is currently unknown if these different types of activation correspond with specific inflammatory subgroups of asthma. We hypothesized that eosinophilic asthma would be characterized by having Th2-associated macrophages, whereas neutrophilic asthma would have Th1-associated macrophages and both having few anti-inflammatory macrophages. We quantified macrophage subsets in bronchial biopsies of asthma patients using interferon regulatory factor 5 (IRF5)/CD68 for Th1-associated macrophages, CD206/CD68 for Th2-associated macrophages and interleukin 10 (IL10)/CD68 for anti-inflammatory macrophages. Macrophage subset percentages were investigated in subgroups of asthma as defined by unsupervised clustering using neutrophil/eosinophil counts in sputum and tissue and forced expiratory volume in 1 s (FEV1). Asthma patients clustered into four subgroups: mixed-eosinophilic/neutrophilic, paucigranulocytic, neutrophilic with normal FEV1, and neutrophilic with low FEV1, the latter group consisting mainly of smokers. No differences were found for CD206+ macrophages within asthma subgroups. In contrast, IRF5+ macrophages were significantly higher and IL10+ macrophages lower in neutrophilic asthmatics with low FEV1 as compared to those with neutrophilic asthma and normal FEV1 or mixed-eosinophilic asthma. This study shows that neutrophilic asthma with low FEV1 is associated with high numbers of IRF5+, and low numbers of IL10+ macrophages, which may be the result of combined effects of smoking and having asthma.
Collapse
Affiliation(s)
- Nil Turan
- GlaxoSmithKline, Allergic Inflammation Discovery Performance Unit, Respiratory Therapy Area, Stevenage, United Kingdom
| | - T. Anienke van der Veen
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, Netherlands
- University Medical Center Groningen, Groningen Research Institute for Asthma and Chronic Obstructive Pulmonary Disease (COPD), University of Groningen, Groningen, Netherlands
| | - Christina Draijer
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, Netherlands
- University Medical Center Groningen, Groningen Research Institute for Asthma and Chronic Obstructive Pulmonary Disease (COPD), University of Groningen, Groningen, Netherlands
| | - Fatemeh Fattahi
- University Medical Center Groningen, Groningen Research Institute for Asthma and Chronic Obstructive Pulmonary Disease (COPD), University of Groningen, Groningen, Netherlands
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Nick H. ten Hacken
- University Medical Center Groningen, Groningen Research Institute for Asthma and Chronic Obstructive Pulmonary Disease (COPD), University of Groningen, Groningen, Netherlands
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Wim Timens
- University Medical Center Groningen, Groningen Research Institute for Asthma and Chronic Obstructive Pulmonary Disease (COPD), University of Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Antoon J. van Oosterhout
- GlaxoSmithKline, Allergic Inflammation Discovery Performance Unit, Respiratory Therapy Area, Stevenage, United Kingdom
| | - Maarten van den Berge
- University Medical Center Groningen, Groningen Research Institute for Asthma and Chronic Obstructive Pulmonary Disease (COPD), University of Groningen, Groningen, Netherlands
- Department of Pulmonology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Barbro N. Melgert
- Department of Molecular Pharmacology, Groningen Research Institute for Pharmacy, University of Groningen, Groningen, Netherlands
- University Medical Center Groningen, Groningen Research Institute for Asthma and Chronic Obstructive Pulmonary Disease (COPD), University of Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- *Correspondence: Barbro N. Melgert
| |
Collapse
|
16
|
Neaga A, Bagacean C, Tempescul A, Jimbu L, Mesaros O, Blag C, Tomuleasa C, Bocsan C, Gaman M, Zdrenghea M. MicroRNAs Associated With a Good Prognosis of Acute Myeloid Leukemia and Their Effect on Macrophage Polarization. Front Immunol 2021; 11:582915. [PMID: 33519805 PMCID: PMC7845488 DOI: 10.3389/fimmu.2020.582915] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/24/2020] [Indexed: 12/24/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive myeloid malignancy with poor outcomes despite very intensive therapeutic approaches. For the majority of patients which are unfit and treated less intensively, the prognosis is even worse. There has been unspectacular progress in outcome improvement over the last decades and the development of new approaches is of tremendous interest. The tumor microenvironment is credited with an important role in supporting cancer growth, including leukemogenesis. Macrophages are part of the tumor microenvironment and their contribution in this setting is increasingly being deciphered, these cells being credited with a tumor supporting role. Data on macrophage role and polarization in leukemia is scarce. MicroRNAs (miRNAs) have a role in the post-transcriptional regulation of gene expression, by impending translation and promoting degradation of messenger RNAs. They are important modulators of cellular pathways, playing major roles in normal hematopoietic differentiation. miRNA expression is significantly correlated with the prognosis of hematopoietic malignancies, including AML. Oncogenic miRNAs correlate with poor prognosis, while tumor suppressor miRNAs, which inhibit the expression of proto-oncogenes, are correlated with a favorable prognosis. miRNAs are proposed as biomarkers for diagnosis and prognosis and are regarded as therapeutic approaches in many cancers, including AML. miRNAs with epigenetic or modulatory activity, as well as with synergistic activity with chemotherapeutic agents, proved to be promising therapeutic targets in experimental, pre-clinical approaches. The clinical availability of emerging compounds with mimicking or suppressor activity provides the opportunity for future therapeutic targeting of miRNAs. The present paper is focusing on miRNAs which, according to current knowledge, favorably impact on AML outcomes, being regarded as tumor suppressors, and reviews their role in macrophage polarization. We are focusing on miRNA expression in the setting of AML, but data on correlations between miRNA expression and macrophage polarization is mostly coming from studies involving normal tissue.
Collapse
Affiliation(s)
- Alexandra Neaga
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Bagacean
- Department of Hematology, Brest University Medical School Hospital, Brest, France.,U1227 B Lymphocytes and Autoimmunity, University of Brest, INSERM, IBSAM, Brest, France
| | - Adrian Tempescul
- Department of Hematology, Brest University Medical School Hospital, Brest, France.,U1227 B Lymphocytes and Autoimmunity, University of Brest, INSERM, IBSAM, Brest, France
| | - Laura Jimbu
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Oana Mesaros
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Cristina Blag
- Department of Pediatrics, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Oncology Institute, Cluj-Napoca, Romania
| | - Corina Bocsan
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihaela Gaman
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.,Department of Hematology, Ion Chiricuta Oncology Institute, Cluj-Napoca, Romania
| |
Collapse
|
17
|
Xu Q, Tang Y, Huang G. Innate immune responses in RNA viral infection. Front Med 2020; 15:333-346. [PMID: 33263837 PMCID: PMC7862985 DOI: 10.1007/s11684-020-0776-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 03/14/2020] [Indexed: 12/17/2022]
Abstract
RNA viruses cause a multitude of human diseases, including several pandemic events in the past century. Upon viral invasion, the innate immune system responds rapidly and plays a key role in activating the adaptive immune system. In the innate immune system, the interactions between pathogen-associated molecular patterns and host pattern recognition receptors activate multiple signaling pathways in immune cells and induce the production of pro-inflammatory cytokines and interferons to elicit antiviral responses. Macrophages, dendritic cells, and natural killer cells are the principal innate immune components that exert antiviral activities. In this review, the current understanding of innate immunity contributing to the restriction of RNA viral infections was briefly summarized. Besides the main role of immune cells in combating viral infection, the intercellular transfer of pathogen and host-derived materials and their epigenetic and metabolic interactions associated with innate immunity was discussed. This knowledge provides an enhanced understanding of the innate immune response to RNA viral infections in general and aids in the preparation for the existing and next emerging viral infections.
Collapse
Affiliation(s)
- Qian Xu
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.,Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuting Tang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Gang Huang
- Divisions of Pathology and Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
| |
Collapse
|
18
|
Wang Y, Wang K, Fu J. HDAC6 Mediates Poly (I:C)-Induced TBK1 and Akt Phosphorylation in Macrophages. Front Immunol 2020; 11:1776. [PMID: 32849638 PMCID: PMC7431618 DOI: 10.3389/fimmu.2020.01776] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/03/2020] [Indexed: 12/21/2022] Open
Abstract
Macrophages are derived from monocytes in the bone marrow and play an important role in anti-viral innate immune responses. Macrophages produce cytokines such as interferons and IL-10 upon viral infection to modulate anti-viral immune responses. Type I interferons (IFNs) promote anti-viral defense. IL-10 is a suppressor cytokine that down-regulates anti-viral immune responses. HDAC6 is a tubulin deacetylase that can modulate microtubule dynamics and microtubule-mediated cell signaling pathways. In the present study, we investigated the potential role of HDAC6 in macrophage anti-viral responses by examining poly (I:C)-induced IFN-β and IL-10 production in mouse bone marrow-derived macrophages (BMDMs). We also investigated the role of HDAC6 in poly (I:C)-induced anti-viral signaling such as TBK1, GSK-3β, and Akt activation in mouse BMDMs. Our data showed that HDAC6 deletion enhanced poly (I:C)-induced INF-β expression in macrophages by up-regulating TBK1 activity and eliminating the inhibitory regulation of GSK-3β. Furthermore, HDAC6 deletion inhibited poly (I:C)-induced suppressor cytokine IL-10 production in the BMDMs, which was associated with the inhibition of Akt activation. Our results suggest that HDAC6 modulates IFN-β and IL-10 production in macrophages through its regulation of TBK1, GSK-3β, and Akt signaling. HDAC6 could act as a suppressor of anti-viral innate immune responses in macrophages.
Collapse
Affiliation(s)
- Yan Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China.,Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Jian Fu
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
19
|
Kinaret PAS, Scala G, Federico A, Sund J, Greco D. Carbon Nanomaterials Promote M1/M2 Macrophage Activation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1907609. [PMID: 32250056 DOI: 10.1002/smll.201907609] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 03/06/2020] [Accepted: 03/08/2020] [Indexed: 05/07/2023]
Abstract
Toxic effects of certain carbon nanomaterials (CNM) have been observed in several exposure scenarios both in vivo and in vitro. However, most of the data currently available has been generated in a high-dose/acute exposure setup, limiting the understanding of their immunomodulatory mechanisms. Here, macrophage-like THP-1 cells, exposed to ten different CNM for 48 h in low-cytotoxic concentration of 10 µg mL-1 , are characterized by secretion of different cytokines and global transcriptional changes. Subsequently, the relationships between cytokine secretion and transcriptional patterns are modeled, highlighting specific pathways related to alternative macrophage activation. Finally, time- and dose-dependent activation of transcription and secretion of M1 marker genes IL-1β and tumor necrosis factor, and M2 marker genes IL-10 and CSF1 is confirmed among the three most responsive CNM, with concentrations of 5, 10, and 20 µg mL-1 at 24, 48, and 72 h of exposure. These results underline CNM effects on the formation of cell microenvironment and gene expression leading to specific patterns of macrophage polarization. Taken together, these findings imply that, instead of a high and toxic CNM dose, a sub-lethal dose in controlled exposure setup can be utilized to alter the cell microenvironment and program antigen presenting cells, with fascinating implications for novel therapeutic strategies.
Collapse
Affiliation(s)
- Pia Anneli Sofia Kinaret
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, 00790, Finland
| | - Giovanni Scala
- Faculty of Biological Sciences, University of Naples, Naples, 80100, Italy
| | - Antonio Federico
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
| | - Jukka Sund
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
| | - Dario Greco
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, 00790, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, 33520, Finland
| |
Collapse
|
20
|
Ramirez R, Herrera AM, Ramirez J, Qian C, Melton DW, Shireman PK, Jin YF. Deriving a Boolean dynamics to reveal macrophage activation with in vitro temporal cytokine expression profiles. BMC Bioinformatics 2019; 20:725. [PMID: 31852428 PMCID: PMC6921543 DOI: 10.1186/s12859-019-3304-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/03/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Macrophages show versatile functions in innate immunity, infectious diseases, and progression of cancers and cardiovascular diseases. These versatile functions of macrophages are conducted by different macrophage phenotypes classified as classically activated macrophages and alternatively activated macrophages due to different stimuli in the complex in vivo cytokine environment. Dissecting the regulation of macrophage activations will have a significant impact on disease progression and therapeutic strategy. Mathematical modeling of macrophage activation can improve the understanding of this biological process through quantitative analysis and provide guidance to facilitate future experimental design. However, few results have been reported for a complete model of macrophage activation patterns. RESULTS We globally searched and reviewed literature for macrophage activation from PubMed databases and screened the published experimental results. Temporal in vitro macrophage cytokine expression profiles from published results were selected to establish Boolean network models for macrophage activation patterns in response to three different stimuli. A combination of modeling methods including clustering, binarization, linear programming (LP), Boolean function determination, and semi-tensor product was applied to establish Boolean networks to quantify three macrophage activation patterns. The structure of the networks was confirmed based on protein-protein-interaction databases, pathway databases, and published experimental results. Computational predictions of the network evolution were compared against real experimental results to validate the effectiveness of the Boolean network models. CONCLUSION Three macrophage activation core evolution maps were established based on the Boolean networks using Matlab. Cytokine signatures of macrophage activation patterns were identified, providing a possible determination of macrophage activations using extracellular cytokine measurements.
Collapse
Affiliation(s)
- Ricardo Ramirez
- Department of Electrical and Computer Engineering, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA
| | - Allen Michael Herrera
- Department of Electrical and Computer Engineering, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA
| | - Joshua Ramirez
- Department of Electrical and Computer Engineering, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA
| | - Chunjiang Qian
- Department of Electrical and Computer Engineering, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA
| | - David W Melton
- Department of Anesthesia, Critical Care and Pain Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
| | - Paula K Shireman
- Department of Surgery, Long School of Medicine, University of Texas Health Science Center San Antonio, 7703 Floyd Curl Dr, San Antonio, TX, 78229, USA
- South Texas Veterans Health Care System, 7400 Merton Minter Blvd, San Antonio, TX, 78229, USA
| | - Yu-Fang Jin
- Department of Electrical and Computer Engineering, The University of Texas at San Antonio, 1 UTSA Circle, San Antonio, TX, 78249, USA.
| |
Collapse
|
21
|
Ferrer MF, Thomas P, López Ortiz AO, Errasti AE, Charo N, Romanowski V, Gorgojo J, Rodriguez ME, Carrera Silva EA, Gómez RM. Junin Virus Triggers Macrophage Activation and Modulates Polarization According to Viral Strain Pathogenicity. Front Immunol 2019; 10:2499. [PMID: 31695702 PMCID: PMC6817498 DOI: 10.3389/fimmu.2019.02499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/07/2019] [Indexed: 01/24/2023] Open
Abstract
The New World arenavirus Junin (JUNV) is the etiological agent of Argentine hemorrhagic fever (AHF). Previous studies of human macrophage infection by the Old-World arenaviruses Mopeia and Lassa showed that while the non-pathogenic Mopeia virus replicates and activates human macrophages, the pathogenic Lassa virus replicates but fails to activate human macrophages. Less is known in regard to the impact of New World arenavirus infection on the human macrophage immune response. Macrophage activation is critical for controlling infections but could also be usurped favoring immune evasion. Therefore, it is crucial to understand how the JUNV infection modulates macrophage plasticity to clarify its role in AHF pathogenesis. With this aim in mind, we compared infection with the attenuated Candid 1 (C#1) or the pathogenic P strains of the JUNV virus in human macrophage cultures. The results showed that both JUNV strains similarly replicated and induced morphological changes as early as 1 day post-infection. However, both strains differentially induced the expression of CD71, the receptor for cell entry, the activation and maturation molecules CD80, CD86, and HLA-DR and selectively modulated cytokine production. Higher levels of TNF-α, IL-10, and IL-12 were detected with C#1 strain, while the P strain induced only higher levels of IL-6. We also found that C#1 strain infection skewed macrophage polarization to M1, whereas the P strain shifted the response to an M2 phenotype. Interestingly, the MERTK receptor, that negatively regulates the immune response, was down-regulated by C#1 strain and up-regulated by P strain infection. Similarly, the target genes of MERTK activation, the cytokine suppressors SOCS1 and SOCS3, were also increased after P strain infection, in addition to IRF-1, that regulates type I IFN levels, which were higher with C#1 compared with P strain infection. Together, this differential activation/polarization pattern of macrophages elicited by P strain suggests a more evasive immune response and may have important implications in the pathogenesis of AHF and underpinning the development of new potential therapeutic strategies.
Collapse
Affiliation(s)
- María F Ferrer
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Pablo Thomas
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Aída O López Ortiz
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina.,Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Andrea E Errasti
- Facultad de Medicina, Instituto de Farmacologia, University of Buenos Aries, Buenos Aires, Argentina
| | - Nancy Charo
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Victor Romanowski
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina.,Global Viral Network, Baltimore, MD, United States
| | - Juan Gorgojo
- Centro de Investigación y Desarrollo en Fermentaciones Industriales, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - María E Rodriguez
- Centro de Investigación y Desarrollo en Fermentaciones Industriales, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Eugenio A Carrera Silva
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Ricardo M Gómez
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina.,Global Viral Network, Baltimore, MD, United States
| |
Collapse
|
22
|
A Modified Collagen Dressing Induces Transition of Inflammatory to Reparative Phenotype of Wound Macrophages. Sci Rep 2019; 9:14293. [PMID: 31586077 PMCID: PMC6778115 DOI: 10.1038/s41598-019-49435-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 08/22/2019] [Indexed: 12/11/2022] Open
Abstract
Collagen containing wound-care dressings are extensively used. However, the mechanism of action of these dressings remain unclear. Earlier studies utilizing a modified collagen gel (MCG) dressing demonstrated improved vascularization of ischemic wounds and better healing outcomes. Wound macrophages are pivotal in facilitating wound angiogenesis and timely healing. The current study was designed to investigate the effect of MCG on wound macrophage phenotype and function. MCG augmented recruitment of macrophage at the wound-site, attenuated pro-inflammatory and promoted anti-inflammatory macrophage polarization. Additionally, MCG increased anti-inflammatory IL-10, IL-4 and pro-angiogenic VEGF production, indicating a direct role of MCG in resolving wound inflammation and improving angiogenesis. At the wound-site, impairment in clearance of apoptotic cell bioburden enables chronic inflammation. Engulfment of apoptotic cells by macrophages (efferocytosis) resolves inflammation via a miR-21-PDCD4-IL-10 pathway. MCG-treated wound macrophages exhibited a significantly bolstered efferocytosis index. Such favorable outcome significantly induced miR-21 expression. MCG-mediated IL-10 production was dampened under conditions of miR-21 knockdown pointing towards miR-21 as a causative factor. Pharmacological inhibition of JNK attenuated IL-10 production by MCG, implicating miR-21-JNK pathway in MCG-mediated IL-10 production by macrophages. This work provides direct evidence demonstrating that a collagen-based wound-care dressing may influence wound macrophage function and therefore modify wound inflammation outcomes.
Collapse
|
23
|
Wang Z, Tan Y, Mou X, Wang C, Li Y, Xiao F, Hu X, Liu H, Xu H. Screening and pharmacodynamic evaluation of the anti-respiratory syncytial virus activity of butene lactones in vitro and in vivo. J Med Virol 2019; 92:17-25. [PMID: 31475735 DOI: 10.1002/jmv.25586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/28/2019] [Indexed: 11/07/2022]
Abstract
A series of butene lactones were synthesized and these compounds were tested for anti-respiratory syncytial virus (RSV) activity in vitro. Three compounds exhibited an antiviral effect, the highest of which was compound 6b3 with an effective concentration 50% of 6.35 μM. The effects of 6b3 were then evaluated in vivo and a significant reduction in the lung index caused by RSV was detected. Reduced inflammatory infiltration and necrosis of the lungs were revealed by histopathology and gross pathology. Activation of an early immune response by 6b3 was also observed by cytokine analysis via a real-time polymerase chain reaction. These results indicated that 6b3 has an anti-RSV effect both in vitro and in vivo, and is a possible candidate compound for the development of an anti-RSV drug in the future.
Collapse
Affiliation(s)
- Zhenya Wang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yayun Tan
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Xiaodong Mou
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan, China
| | - Congcong Wang
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yuanyuan Li
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan, China
| | - Fan Xiao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Xiaoning Hu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Hongmin Liu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Haiwei Xu
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory of "Runliang" Antiviral Medicines Research and Development, Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Fox KA, Kirwan DE, Whittington AM, Krishnan N, Robertson BD, Gilman RH, López JW, Singh S, Porter JC, Friedland JS. Platelets Regulate Pulmonary Inflammation and Tissue Destruction in Tuberculosis. Am J Respir Crit Care Med 2019; 198:245-255. [PMID: 29420060 DOI: 10.1164/rccm.201710-2102oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
RATIONALE Platelets may interact with the immune system in tuberculosis (TB) to regulate human inflammatory responses that lead to morbidity and spread of infection. OBJECTIVES To identify a functional role of platelets in the innate inflammatory and matrix-degrading response in TB. METHODS Markers of platelet activation were examined in plasma from 50 patients with TB before treatment and 50 control subjects. Twenty-five patients were followed longitudinally. Platelet-monocyte interactions were studied in a coculture model infected with live, virulent Mycobacterium tuberculosis (M.tb) and dissected using qRT-PCR, Luminex multiplex arrays, matrix degradation assays, and colony counts. Immunohistochemistry detected CD41 (cluster of differentiation 41) expression in a pulmonary TB murine model, and secreted platelet factors were measured in BAL fluid from 15 patients with TB and matched control subjects. MEASUREMENTS AND MAIN RESULTS Five of six platelet-associated mediators were upregulated in plasma of patients with TB compared with control subjects, with concentrations returning to baseline by Day 60 of treatment. Gene expression of the monocyte collagenase MMP-1 (matrix metalloproteinase-1) was upregulated by platelets in M.tb infection. Platelets also enhanced M.tb-induced MMP-1 and -10 secretion, which drove type I collagen degradation. Platelets increased monocyte IL-1 and IL-10 and decreased IL-12 and MDC (monocyte-derived chemokine; also known as CCL-22) secretion, as consistent with an M2 monocyte phenotype. Monocyte killing of intracellular M.tb was decreased. In the lung, platelets were detected in a TB mouse model, and secreted platelet mediators were upregulated in human BAL fluid and correlated with MMP and IL-1β concentrations. CONCLUSIONS Platelets drive a proinflammatory, tissue-degrading phenotype in TB.
Collapse
Affiliation(s)
- Katharine A Fox
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Daniela E Kirwan
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Ashley M Whittington
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Nitya Krishnan
- 2 Medical Research Council Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London, United Kingdom
| | - Brian D Robertson
- 2 Medical Research Council Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London, United Kingdom
| | - Robert H Gilman
- 3 Department of International Health, Johns Hopkins University, Baltimore Maryland.,4 Laboratorio de Investigación en Enfermedades Infecciosas, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - José W López
- 5 Laboratorio de Bioinformática y Biología Molecular, Universidad Peruana Cayetano Heredia, Lima, Peru.,6 Instituto Nacional de Salud del Niño, Lima, Peru; and
| | - Shivani Singh
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Joanna C Porter
- 7 Centre for Inflammation and Tissue Repair, Respiratory Medicine, University College London, United Kingdom
| | - Jon S Friedland
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| |
Collapse
|
25
|
Feketea G, Bocsan CI, Popescu C, Gaman M, Stanciu LA, Zdrenghea MT. A Review of Macrophage MicroRNAs' Role in Human Asthma. Cells 2019; 8:cells8050420. [PMID: 31071965 PMCID: PMC6562863 DOI: 10.3390/cells8050420] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
There is an imbalance in asthma between classically activated macrophages (M1 cells) and alternatively activated macrophages (M2 cells) in favor of the latter. MicroRNAs (miRNAs) play a critical role in regulating macrophage proliferation and differentiation and control the balance of M1 and M2 macrophage polarization, thereby controlling immune responses. Here we review the current published data concerning miRNAs with known correlation to a specific human macrophage phenotype and polarization, and their association with adult asthma. MiRNA-targeted therapy is still in the initial stages, but clinical trials are under recruitment or currently running for some miRNAs in other diseases. Regulating miRNA expression via their upregulation or downregulation could show potential as a novel therapy for improving treatment efficacy in asthma.
Collapse
Affiliation(s)
- Gavriela Feketea
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania.
| | - Corina I Bocsan
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.
| | - Cristian Popescu
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania.
| | - Mihaela Gaman
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| | - Luminita A Stanciu
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK.
| | - Mihnea T Zdrenghea
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania.
- Department of Hematology, Ion Chiricuta Oncology Institute, 400010 Cluj-Napoca, Romania.
| |
Collapse
|
26
|
Localized Osteoarthritis Disease-Modifying Changes due to Intra-articular Injection of Micronized Dehydrated Human Amnion/Chorion Membrane. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-018-0087-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
27
|
El Masry MS, Chaffee S, Das Ghatak P, Mathew-Steiner SS, Das A, Higuita-Castro N, Roy S, Anani RA, Sen CK. Stabilized collagen matrix dressing improves wound macrophage function and epithelialization. FASEB J 2019; 33:2144-2155. [PMID: 30260708 PMCID: PMC6338656 DOI: 10.1096/fj.201800352r] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022]
Abstract
Decellularized matrices of biologic tissue have performed well as wound care dressings. Extracellular matrix-based dressings are subject to rapid degradation by excessive protease activity at the wound environment. Stabilized, acellular, equine pericardial collagen matrix (sPCM) wound care dressing is flexible cross-linked proteolytic enzyme degradation resistant. sPCM was structurally characterized utilizing scanning electron and atomic force microscopy. In murine excisional wounds, sPCM was effective in mounting an acute inflammatory response. Postwound inflammation resolved rapidly, as indicated by elevated levels of IL-10, arginase-1, and VEGF, and lowering of IL-1β and TNF-α. sPCM induced antimicrobial proteins S100A9 and β-defensin-1 in keratinocytes. Adherence of Pseudomonas aeruginosa and Staphylococcus aureus on sPCM pre-exposed to host immune cells in vivo was inhibited. Excisional wounds dressed with sPCM showed complete closure at d 14, while control wounds remained open. sPCM accelerated wound re-epithelialization. sPCM not only accelerated wound closure but also improved the quality of healing by increased collagen deposition and maturation. Thus, sPCM is capable of presenting scaffold functionality during the course of wound healing. In addition to inducing endogenous antimicrobial defense systems, the dressing itself has properties that minimize biofilm formation. It mounts robust inflammation, a process that rapidly resolves, making way for wound healing to advance.-El Masry, M. S., Chaffee, S., Das Ghatak, P., Mathew-Steiner, S. S., Das, A., Higuita-Castro, N., Roy, S., Anani, R. A., Sen, C. K. Stabilized collagen matrix dressing improves wound macrophage function and epithelialization.
Collapse
Affiliation(s)
- Mohamed S. El Masry
- Department of Surgery, Indiana University Health (IUH) Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
- Department of Plastic and Reconstructive Surgery, Zagazig University, Zagazig, Egypt
| | - Scott Chaffee
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Piya Das Ghatak
- Department of Surgery, Indiana University Health (IUH) Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Shomita S. Mathew-Steiner
- Department of Surgery, Indiana University Health (IUH) Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Amitava Das
- Department of Surgery, Indiana University Health (IUH) Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Natalia Higuita-Castro
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Sashwati Roy
- Department of Surgery, Indiana University Health (IUH) Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| | - Raafat A. Anani
- Department of Plastic and Reconstructive Surgery, Zagazig University, Zagazig, Egypt
| | - Chandan K. Sen
- Department of Surgery, Indiana University Health (IUH) Comprehensive Wound Center, Indiana Center for Regenerative Medicine and Engineering, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Surgery, The Ohio State University, Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
28
|
Miliani M, Nouar M, Paris O, Lefranc G, Mennechet F, Aribi M. Thymoquinone Potently Enhances the Activities of Classically Activated Macrophages Pulsed with Necrotic Jurkat Cell Lysates and the Production of Antitumor Th1-/M1-Related Cytokines. J Interferon Cytokine Res 2018; 38:539-551. [PMID: 30422744 DOI: 10.1089/jir.2018.0010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Antitumor activity of classically activated macrophage (Mϕ) may be impaired within the tumors, spleen, and bone marrow. Thus, it is possible to boost its antitumor activity after its pulsing with necrotic tumor cell lysates combined with an adjuvant. We set out to determine the potential adjuvant effects of thymoquinone (TQ; 2-isopropyl-5-methyl-1,4-benzoquinone, C10H12O2) on both functional activities of classically activated Mϕs, pulsed or not with necrotic Jurkat T cell line lysates (NecrJCL), and the balance of antitumor cytokines (ATCs) versus immunosuppressive cytokines (ISCs) during crosstalk with autologous human CD4+ T cells. We found that TQ treatment resulted in a significant upregulation of phagocytic activity, respiratory burst, the production of interleukin-2 (IL-2), IL-6, and IL-17 in NecrJCL-pulsed Mϕ co-culture system, and, conversely, in downregulation of the production of IL-6, IL-17, nitric oxide (NO), and arginase activity in nonpulsed TQ-treated Mϕs co-culture system. In addition, TQ has also shown low upregulation effect on the production of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and IL-1β, pathogen killing capacity and H2O2 in NecrJCL-pulsed Mϕs co-cultures. Moreover, TQ significantly downregulated arginase activity, and significantly upregulated inducible NO synthase (iNOS) activity-to-arginase activity ratio in NecrJCL-pulsed Mϕ co-cultures. Furthermore, TQ downregulated IL-10-to-IL-17 ratio and total cellular cholesterol content (ttcCHOL), but upregulated the ratios of IL-1β-to-IL-4, IL-1β-to-IL-10, IFN-γ-to-IL-4, IFN-γ-to-IL-10, TNF-α-to-IL-4, TNF-α-to-IL-10, and combined proinflammatory cytokines (PICs)-to-anti-inflammatory cytokines (AICs) in NecrJCL-pulsed Mϕs co-culture system, whereas significant differences were highlighted only for IL-10-to-IL-17, IFN-γ-to-IL-10, and PICs-to-AICs ratios. Our outcomes demonstrated that TQ can act as potent adjuvant for enhancing both the functional activities of NecrJCL-pulsed Mϕ and the production of ATCs during their interplay with CD4+ T cells.
Collapse
Affiliation(s)
- Maroua Miliani
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Mouna Nouar
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Océane Paris
- Institut de Génétique Moléculaire de Montpellier (IGMM)-UMR5535, CNRS et Université de Montpellier, Montpellier, France
| | - Gérard Lefranc
- Institut de Génétique Humaine, UMR 9002 CNRS-Université de Montpellier, Montpellier, France
| | - Franck Mennechet
- Institut de Génétique Moléculaire de Montpellier (IGMM)-UMR5535, CNRS et Université de Montpellier, Montpellier, France
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| |
Collapse
|
29
|
Wang P, Xu LJ, Qin JJ, Zhang L, Zhuang GH. MicroRNA-155 inversely correlates with esophageal cancer progression through regulating tumor-associated macrophage FGF2 expression. Biochem Biophys Res Commun 2018; 503:452-458. [PMID: 29660336 DOI: 10.1016/j.bbrc.2018.04.094] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/12/2018] [Indexed: 11/26/2022]
Abstract
Esophageal cancer (EC) is one of the most common malignancies with high incidence and mortality. Tumor-associated macrophages (TAMs) in the tumor microenvironment have been linked to the accelerated tumor progression. MicroRNAs (miR) are 19-25 nucleotide-long, noncoding RNA molecules, functioning as modulators of gene expression, and mediate a variety of biological functions, including tumor growth. In the present study, the effects and molecular mechanism of miR-155 in TAMs isolated from EC were explored. The expression of miR-155 and fibroblast growth factor-2 (FGF2) in EC tissues and cell lines were analyzed using reverse transcription-quantitative PCR (qRT-PCR) and western blot assays. TAMs were also transfected with the described constructs. Following, the culture medium from TAMs was collected for further analysis. The released FGF2, and inflammatory cytokines were quantified using ELISA. The cell viability, migrated and invaded levels were calculated through Cell Counting kit-8 (CCK8), and transwell analysis. Moreover, human umbilical vein endothelial cells (HUVEC) vasculature formation was determined using matrigel angiogenesis analysis. The results indicated that miR-155 expression was decreased in EC tissues and cell lines, while FGF2 expression was increased in comparison to those in the normal control group. Moreover, miR-155 mimics transfection up-regulated tumor necrosis factor α (TNF-α), interleukin (IL)-12 and inducible nitric oxide synthase (iNOS), while down-regulated IL-10, Arginase-1 (Arg-1) and IL-22 levels in the culture medium from TAMs. And enhancing miR-155 expression in TAMs suppressed the cell viability, migration and invasion of ECA109 cells and reduced the angiogenesis. Nevertheless, over-expressing FGF2 abolished the role of miR-155 in cancer cell survival, migration, invasion as well as angiogenesis. Our findings indicated that miR-155-regulated FGF2 expression from TAMs suppressed EC cell proliferation, migration, invasion and inhibited vasculature formation. Thus, miR-155-modulated FGF2 might be a potential therapeutic target to prevent EC progression.
Collapse
Affiliation(s)
- Peng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiao Tong University Health Science Center, Xi'an, Shaanxi 710061, China; College of Public Health and Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Li-Juan Xu
- Department of Clinical Laboratory, The First Affiliated Hospital, Zhengzhou University, Zhengzhou 450052, China
| | - Jie-Jie Qin
- College of Public Health and Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Lu Zhang
- College of Public Health and Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Gui-Hua Zhuang
- Department of Epidemiology and Biostatistics, School of Public Health, Xi'an Jiao Tong University Health Science Center, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
30
|
Qiu ZQ, Han B, Zhang ZQ, Wang X, Li LS, Xu JD. Biological characteristics of intestinal IgE and gut diseases. Shijie Huaren Xiaohua Zazhi 2018; 26:110-119. [DOI: 10.11569/wcjd.v26.i2.110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Immunoglobulin E (IgE), a crucial protective substance for the intestinal tract, plays an important role in gut immunity. IgE is secreted by plasma cells in the submucosal lamina propria upon antigenic invasion and, together with certain cytokines and immune cells, is involved in the regulation of gastrointestinal immunity in normal or abnormal conditions via the high affinity IgE receptor (FcεR I) and low affinity IgE receptor (CD23+). In this paper, we review the structure, synthetic transport, secretory regulation, receptor classification, and function of intestinal IgE as well as the related gut diseases.
Collapse
Affiliation(s)
- Zhi-Qiang Qiu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Bo Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Zi-Qing Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xue Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Li-Sheng Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Jing-Dong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| |
Collapse
|
31
|
Roh DE, Park SH, Choi HJ, Kim YH. Comparison of cytokine expression profiles in infants with a rhinovirus induced lower respiratory tract infection with or without wheezing: a comparison with respiratory syncytial virus. KOREAN JOURNAL OF PEDIATRICS 2017; 60:296-301. [PMID: 29042873 PMCID: PMC5638836 DOI: 10.3345/kjp.2017.60.9.296] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 07/10/2017] [Accepted: 08/07/2017] [Indexed: 01/14/2023]
Abstract
PURPOSE The aim of this study was to evaluate whether infants with rhinovirus (RV) infection-induced wheezing and those with respiratory syncytial virus (RSV) infection-induced wheezing have different cytokine profiles in the acute stage. METHODS Of the infants with lower respiratory tract infection (LRTI) between September 2011 and May 2012, 88 were confirmed using reverse transcription polymerase chain reaction and hospitalized. Systemic interferon-gamma (IFN-γ), interleukin (IL)-2, IL-12, IL-4, IL-5, IL-13, and Treg-type cytokine (IL-10) responses were examined with multiplex assay using acute phase serum samples. RESULTS Of the 88 patients, 38 had an RV infection (RV group) and 50 had an RSV infection (RSV group). In the RV group, the IFN-γ and IL-10 concentrations were higher in the patients with than in the patients without wheezing (P=0.022 and P=0.007, respectively). In the RSV group, the differences in IFN-γ and IL-10 concentrations did not reach statistical significance between the patients with and the patients without wheezing (P=0.105 and P=0.965, respectively). The IFN-γ and IL-10 concentrations were not significantly different between the RV group with wheezing and the RSV group with wheezing (P=0.155 and P=0.801, respectively), in contrast to the significant difference between the RV group without wheezing and the RSV group without wheezing (P=0.019 and P=0.035, respectively). CONCLUSION In comparison with RSV-induced LRTI, RV-induced LRTI combined with wheezing showed similar IFN-γ and IL-10 levels, which may have an important regulatory function.
Collapse
Affiliation(s)
- Da Eun Roh
- Department of Pediatrics, Kyungpook National University School of Medicine, Daegu, Korea
| | - Sook-Hyun Park
- Department of Pediatrics, Kyungpook National University School of Medicine, Daegu, Korea
| | - Hee Joung Choi
- Department of Pediatrics, Keimyung University School of Medicine, Daegu, Korea
| | - Yeo Hyang Kim
- Department of Pediatrics, Kyungpook National University School of Medicine, Daegu, Korea
| |
Collapse
|
32
|
Upton N, Jackson DJ, Nikonova AA, Hingley-Wilson S, Khaitov M, del Rosario A, Traub S, Trujillo-Torralbo MB, Habibi M, Elkin SL, Kon OM, Edwards MR, Mallia P, Footitt J, Macintyre J, Stanciu LA, Johnston SL, Sykes A. Rhinovirus induction of fractalkine (CX3CL1) in airway and peripheral blood mononuclear cells in asthma. PLoS One 2017; 12:e0183864. [PMID: 28859129 PMCID: PMC5578648 DOI: 10.1371/journal.pone.0183864] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 08/12/2017] [Indexed: 12/21/2022] Open
Abstract
Rhinovirus infection is associated with the majority of asthma exacerbations. The role of fractalkine in anti-viral (type 1) and pathogenic (type 2) responses to rhinovirus infection in allergic asthma is unknown. To determine whether (1) fractalkine is produced in airway cells and in peripheral blood leucocytes, (2) rhinovirus infection increases production of fractalkine and (3) levels of fractalkine differ in asthmatic compared to non-asthmatic subjects. Fractalkine protein and mRNA levels were measured in bronchoalveolar lavage (BAL) cells and peripheral blood mononuclear cells (PBMCs) from non-asthmatic controls (n = 15) and mild allergic asthmatic (n = 15) subjects. Protein levels of fractalkine were also measured in macrophages polarised ex vivo to give M1 (type 1) and M2 (type 2) macrophages and in BAL fluid obtained from mild (n = 11) and moderate (n = 14) allergic asthmatic and non-asthmatic control (n = 10) subjects pre and post in vivo rhinovirus infection. BAL cells produced significantly greater levels of fractalkine than PBMCs. Rhinovirus infection increased production of fractalkine by BAL cells from non-asthmatic controls (P<0.01) and in M1-polarised macrophages (P<0.05), but not in BAL cells from mild asthmatics or in M2 polarised macrophages. Rhinovirus induced fractalkine in PBMCs from asthmatic (P<0.001) and healthy control subjects (P<0.05). Trends towards induction of fractalkine in moderate asthmatic subjects during in vivo rhinovirus infection failed to reach statistical significance. Fractalkine may be involved in both immunopathological and anti-viral immune responses to rhinovirus infection. Further investigation into how fractalkine is regulated across different cell types and into the effect of stimulation including rhinovirus infection is warranted to better understand the precise role of this unique dual adhesion factor and chemokine in immune cell recruitment.
Collapse
Affiliation(s)
- Nadine Upton
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Randall Division of Cell and Molecular Biophysics, Kings College London, London, United Kingdom
| | - David J. Jackson
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Alexandra A. Nikonova
- NRC institute of Immunology FMBA, Moscow, Russian Federation
- Mechnikov Research Institute for Vaccines and Sera, Moscow, Russian Federation
- * E-mail:
| | - Suzie Hingley-Wilson
- Respiratory Infection Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Musa Khaitov
- NRC institute of Immunology FMBA, Moscow, Russian Federation
| | - Ajerico del Rosario
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Stephanie Traub
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Maria-Belen Trujillo-Torralbo
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Max Habibi
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
- Respiratory Infection Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Sarah L. Elkin
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Onn M. Kon
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Michael R. Edwards
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Patrick Mallia
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Joseph Footitt
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Jonathan Macintyre
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Luminita A. Stanciu
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
| | - Sebastian L. Johnston
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Annemarie Sykes
- Airway Disease Infection Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, London, United Kingdom
- Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
33
|
OM-85 is an immunomodulator of interferon-β production and inflammasome activity. Sci Rep 2017; 7:43844. [PMID: 28262817 PMCID: PMC5338315 DOI: 10.1038/srep43844] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/30/2017] [Indexed: 12/12/2022] Open
Abstract
The inflammasome–IL-1 axis and type I interferons (IFNs) have been shown to exert protective effects upon respiratory tract infections. Conversely, IL-1 has also been implicated in inflammatory airway pathologies such as asthma and chronic obstructive pulmonary disease (COPD). OM-85 is a bacterial extract with proved efficacy against COPD and recurrent respiratory tract infections, a cause of co-morbidity in asthmatic patients. We therefore asked whether OM-85 affects the above-mentioned innate immune pathways. Here we show that OM-85 induced interferon-β through the Toll-like receptor adaptors Trif and MyD88 in bone marrow-derived dendritic cells. Moreover, it exerted a dual role on IL-1 production; on the one hand, it upregulated proIL-1β and proIL-1α levels in a MyD88-dependent manner without activating the inflammasome. On the other hand, it repressed IL-1β secretion induced by alum, a well-known NLRP3 activator. In vivo, OM-85 diminished the recruitment of inflammatory cells in response to peritoneal alum challenge. Our findings therefore suggest that OM-85 favors a protective primed state, while dampening inflammasome activation in specific conditions. Taken together, these data bring new insights into the mechanisms of OM-85 action on innate immune pathways and suggest potential explanations for its efficacy in the treatment of virus-induced airway diseases.
Collapse
|
34
|
Yang Y, Wu BQ, Wang YH, Shi YF, Luo JM, Ba JH, Liu H, Zhang TT. Regulatory effects of miR-155 and miR-146a on repolarization and inflammatory cytokine secretion in human alveolar macrophages in vitro. Immunopharmacol Immunotoxicol 2016; 38:502-509. [DOI: 10.1080/08923973.2016.1248845] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Yang Yang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Ben-Quan Wu
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Yan-Hong Wang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Yun-Feng Shi
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Jin-Mei Luo
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Jun-Hui Ba
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Hui Liu
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| | - Tian-Tuo Zhang
- Department of Internal Medicine, Medical Intensive Care Unit and Division of Respiratory Diseases, Institute of Respiratory Disease and the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, People’s Republic of China
| |
Collapse
|
35
|
Geng XR, Qiu SQ, Yang LT, Liu ZQ, Yang G, Liu JQ, Zeng L, Li XX, Mo LH, Liu ZG, Yang PC. Allergen-specific immune response suppresses interleukin 10 expression in B cells via increasing micro-RNA-17-92 cluster. Cell Biochem Funct 2016; 34:449-54. [PMID: 27491928 DOI: 10.1002/cbf.3207] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Xiao-Rui Geng
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
- Longgang ENT Hospital; Shenzhen China
| | - Shu-Qi Qiu
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
- Longgang ENT Hospital; Shenzhen China
| | - Li-Tao Yang
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
- Longgang ENT Hospital; Shenzhen China
| | - Zhi-Qiang Liu
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
- Longgang ENT Hospital; Shenzhen China
| | - Gui Yang
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
- Longgang ENT Hospital; Shenzhen China
| | - Jiang-Qi Liu
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
- Longgang ENT Hospital; Shenzhen China
| | - Lu Zeng
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
| | - Xiao-Xi Li
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
| | - Li-Hua Mo
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
- Longgang ENT Hospital; Shenzhen China
| | - Zhi-Gang Liu
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
| | - Ping-Chang Yang
- ENT Institute of the Research Center of Allergy & Immunology; Shenzhen University School of Medicine; Shenzhen China
| |
Collapse
|
36
|
Mesenchymal Stem Cells and Myeloid Derived Suppressor Cells: Common Traits in Immune Regulation. J Immunol Res 2016; 2016:7121580. [PMID: 27529074 PMCID: PMC4978836 DOI: 10.1155/2016/7121580] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/08/2016] [Indexed: 02/08/2023] Open
Abstract
To protect host against immune-mediated damage, immune responses are tightly regulated. The regulation of immune responses is mediated by various populations of mature immune cells, such as T regulatory cells and B regulatory cells, but also by immature cells of different origins. In this review, we discuss regulatory properties and mechanisms whereby two distinct populations of immature cells, mesenchymal stem cells, and myeloid derived suppressor cells mediate immune regulation, focusing on their similarities, discrepancies, and potential clinical applications.
Collapse
|
37
|
Abstract
Lung macrophages link innate and adaptive immune responses during allergic airway inflammatory responses. Alveolar macrophages (AMs) and interstitial macrophages are two different phenotypes that differentially exert immunological function under physiological and pathological conditions. Exposure to pathogen induces polarization of AM cells into classically activated macrophages (M1 cells) and alternatively activated macrophages (M2 cells). M1 cells dominantly express proinflammatory cytokines such as TNF-α and IL-1 β and induce lung inflammation and tissue damage. M2 cells are further divided into M2a and M2c subsets. M2a cells dominantly produce allergic cytokines IL-4 and IL-13, but M2c cells dominantly produce anti-inflammatory cytokine IL-10. M2a and M2c cells are differently involved in initiation, inflammation resolution, and tissue remodeling in the different stages of asthma. Microenvironment dynamically influences polarization of AM cells. Cytokines, chemokines, and immune-regulatory cells interplay and affect the balance between the polarization of M1 and M2 cells, subsequently influencing disease progression. Thus, modulation of AM phenotypes through molecular intervention has therapeutic potential in the treatment of asthma and other allergic inflammatory diseases. This review updated recent advances in polarization and functional specialization of these macrophage subtypes with emphasis on modulation of polarization of M2 cells in asthma of human subjects and animal models.
Collapse
Affiliation(s)
- Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Lei Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
38
|
Insight into Reepithelialization: How Do Mesenchymal Stem Cells Perform? Stem Cells Int 2015; 2016:6120173. [PMID: 26770209 PMCID: PMC4684897 DOI: 10.1155/2016/6120173] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/01/2015] [Accepted: 07/22/2015] [Indexed: 12/11/2022] Open
Abstract
Wound reepithelialization is a cooperative multifactorial process dominated by keratinocyte migration, proliferation, and differentiation that restores the intact epidermal barrier to prevent infection and excessive moisture loss. However, in wounds that exhibit impaired wound healing, such as chronic nonhealing wounds or hypertrophic scars, the reepithelialization process has failed. Thus, it is necessary to explore a suitable way to mitigate these abnormalities to promote reepithelialization and achieve wound healing. Mesenchymal stem cells (MSCs) have the capacity for self-renewal as well as potential multipotency. These cells play important roles in many biological processes, including anti-inflammation, cell migration, proliferation, and differentiation, and signal pathway activation or inhibition. The mechanism of the involvement of MSCs in reepithelialization is still not fully understood. An abundance of evidence has shown that MSCs participate in reepithelialization by inhibiting excessive inflammatory responses, secreting important factors, differentiating into multiple skin cell types, and recruiting other host cells. This review describes the evidence for the roles that MSCs appear to play in the reepithelialization process.
Collapse
|
39
|
Jakeman PG, Hills TE, Fisher KD, Seymour LW. Macrophages and their interactions with oncolytic viruses. Curr Opin Pharmacol 2015; 24:23-9. [PMID: 26164569 DOI: 10.1016/j.coph.2015.06.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/21/2015] [Accepted: 06/23/2015] [Indexed: 11/24/2022]
Abstract
Macrophages are a highly plastic cell type and exhibit a range of defensive and regulatory functions in normal physiology. Phagocytic macrophages play an important role in defending against virus infection and they provide an important barrier that can limit the delivery of therapeutic viruses from the injection to the tumour. Within tumours, macrophages generally adopt an immunosuppressive phenotype and are associated with poor clinical prognosis. However their plasticity also provides the opportunity for therapeutic 're-education' of tumour-associated macrophages (TAMs) to adopt an active anticancer role. Oncolytic viruses present the possibility for non-specific stimulation of TAMs, and also the option for tumour-targeted expression of cytokines chosen specifically to modulate macrophage activation.
Collapse
Affiliation(s)
| | - Thomas E Hills
- Department of Oncology, University of Oxford, OX3 7DQ, UK
| | - Kerry D Fisher
- Department of Oncology, University of Oxford, OX3 7DQ, UK
| | | |
Collapse
|
40
|
Abstract
Macrophage involvement in viral infections and antiviral states is common. However, this involvement has not been well-studied in the paradigm of macrophage polarization, which typically has been categorized by the dichotomy of classical (M1) and alternative (M2) statuses. Recent studies have revealed the complexity of macrophage polarization in response to various cellular mediators and exogenous stimuli by adopting a multipolar view to revisit the differential process of macrophages, especially those re-polarized during viral infections. Here, through examination of viral infections targeting macrophages/monocytic cells, we focus on the direct involvement of macrophage polarization during viral infections. Type I and type III interferons (IFNs) are critical in regulation of viral pathogenesis and host antiviral infection; thus, we propose to incorporate IFN-mediated antiviral states into the framework of macrophage polarization. This view is supported by the multifunctional properties of type I IFNs, which potentially elicit and regulate both M1- and M2-polarization in addition to inducing the antiviral state, and by the discoveries of viral mechanisms to adapt and modulate macrophage polarization. Indeed, several recent studies have demonstrated effective prevention of viral diseases through manipulation of macrophage immune statuses.
Collapse
Affiliation(s)
- Yongming Sang
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Laura C Miller
- Virus and Prion Diseases of Livestock Research Unit, National Animal Disease Center, USDA-ARS, 1920 Dayton Ave, Ames, IA 50010, USA
| | - Frank Blecha
- Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|