1
|
Dinh KM, Kaspersen KA, Mikkelsen S, Kjerulff BD, Boldsen JK, Petersen MS, Burgdorf KS, Sørensen E, Aagaard B, Forman-Ankjær B, Bruun MT, Banasik K, Hansen TF, Nyegaard M, Rohde PD, Brunak S, Hjalgrim H, Ostrowski SR, Pedersen OB, Ullum H, Erikstrup LT, Erikstrup C. Impact of CCR5Δ32 on the risk of infection, Staphylococcus aureus carriage, and plasma concentrations of chemokines in Danish blood donors. EBioMedicine 2024; 109:105406. [PMID: 39437658 PMCID: PMC11536029 DOI: 10.1016/j.ebiom.2024.105406] [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: 03/01/2024] [Revised: 10/03/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND The CC chemokine receptor 5 (CCR5) is a suggested receptor for Staphylococcus aureus leukotoxin ED. Homozygosity for the Δ32 deletion (CCR5Δ32) protects against human immunodeficiency virus infection and possibly also against leukotoxin ED. We examined the impact of CCR5Δ32 on the susceptibility to S. aureus infection, all-cause infections, and S. aureus nasal carriage, respectively, and on the concentrations of circulating chemokines in blood donors. METHODS We included 95,406 participants from the Danish Blood Donor Study (DBDS) genotyped for >650,000 single nucleotide polymorphisms. The CCR5Δ32 (rs333, MAF: 0.12) was imputed from a reference panel and validated. Infectious outcomes were identified by diagnosis codes and redeemed prescription of antibiotics in national health registers. Data on S. aureus nasal carriage and forty-seven inflammatory biomarkers were available for 6721 and 7811 participants, respectively. Cox, logistic, and linear regression models adjusted for relevant confounders were used to explore said associations. FINDINGS During more than 700,000 person-years of observation, we found that CCR5Δ32 was associated with neither an increased risk of redeemed dicloxacillin, hospital-treated S. aureus-associated infection (replicated in 345,996 Icelanders), redeemed antibiotics, all-cause infection, and nor with S. aureus nasal carriage. We discovered an association between CCR5Δ32 and elevated CCL4 concentrations, which were 1.26-fold higher in Δ32-heterozygotes (95%-CI: 1.23-1.30) and 2.64-fold higher in Δ32-homozygotes (95%-CI: 2.41-2.90) compared with wildtype homozygotes. Conversely, concentrations of CCL2, CXCL-10, and CCL11 were slightly lower among Δ32-heterozygotes. INTERPRETATION Results from this CCR5Δ32 high-prevalent cohort do not support the idea that CCR5Δ32 affects the risk of S. aureus carriage or infection to any relevant degree, in this northern European context. CCL4 was the main chemokine affected by CCR5Δ32 and was observed in higher concentration among Δ32-carriers. This study cannot rule out that S. aureus is a previous driver of CCR5Δ32 selection. FUNDING The Health Research Fund of Central Denmark Region, Aarhus University, Danish Administrative Regions, Bio- and Genome Bank Denmark, Danish Blood Donor Research Foundation, Aase & Ejnar Danielsens Foundation, Højmosegård Grant, National Institute of Allergy and Infectious Diseases, and A.P. Møller Foundation for the Advancement of Medical Science.
Collapse
Affiliation(s)
- Khoa Manh Dinh
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Kathrine Agergård Kaspersen
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark; Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Aarhus, Denmark
| | - Susan Mikkelsen
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Bertram Dalskov Kjerulff
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark; Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Aarhus, Denmark
| | - Jens Kjærgaard Boldsen
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark; Danish Big Data Centre for Environment and Health (BERTHA), Aarhus University, Aarhus, Denmark
| | | | | | - Erik Sørensen
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Bitten Aagaard
- Department of Clinical Immunology, Aalborg University Hospital, Aalborg, Denmark
| | | | - Mie Topholm Bruun
- Department of Clinical Immunology, Odense University Hospital, Odense, Denmark
| | - Karina Banasik
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Folkmann Hansen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Danish Headache Center, Department of Neurology, Copenhagen University Hospital, Glostrup, Denmark
| | - Mette Nyegaard
- Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Palle Duun Rohde
- Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Hjalgrim
- Danish Cancer Society Research Center, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Epidemiology Research, Statens Serum Institut; Department of Haematology, Copenhagen University Hospital, Rigshospitalet
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ole Birger Pedersen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
| | | | | | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
2
|
Liu X, Ji JX, Pang AN, Li L, Nie P, Zhang LQ, Zeng KW, Chen SN. Molecular cloning and functional analyses of C-C motif chemokine ligand 3 (CCL3) in mandarin fish Siniperca chuatsi. FISH & SHELLFISH IMMUNOLOGY 2024; 149:109614. [PMID: 38710342 DOI: 10.1016/j.fsi.2024.109614] [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: 01/21/2024] [Revised: 04/29/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Chemokines are critical molecules involved in immune reaction and immune system homeostasis, and some chemokines play a role in antiviral immunity. It is not known if the C-C motif chemokine ligand 3 (CCL3), a member of the CC chemokine family, possesses antiviral properties in fish. In this study, a ccl3 was cloned from the mandarin fish (Siniperca chuatsi), and it has an open reading frame (ORF) of 276 base pairs, which are predicted to encode a 91-amino acid peptide. Mandarin fish CCL3 revealed conserved sequence features with four cysteine residues and closely relationships with the CCL3s from other vertebrates based on the sequence alignment and phylogenetic analysis. The transcripts of ccl3 were notably enriched in immune-related organs, such as spleen and gills in healthy mandarin fish, and the ccl3 was induced in the isolated mandarin fish brain (MFB) cells following infection with infectious spleen and kidney necrosis virus (ISKNV). Moreover, in MFB cells, overexpression of CCL3 induced immune factors, such as IL1β, TNFα, MX, IRF1 and IFNh, and exhibited antiviral activity against ISKNV. This study sheds light on the immune role of CCL3 in immune response of mandarin fish, and its antiviral defense mechanism is of interest for further investigation.
Collapse
Affiliation(s)
- Xiao Liu
- Wuhan Fisheries Science Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, Hubei Province, 430070, China; College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei Province, 430070, China
| | - Jia Xiang Ji
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - An Ning Pang
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Li Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Li Qiang Zhang
- Wuhan Fisheries Science Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, Hubei Province, 430070, China
| | - Ke Wei Zeng
- Wuhan Fisheries Science Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, Hubei Province, 430070, China; College of Fisheries, Huazhong Agricultural University, Wuhan, Hubei Province, 430070, China.
| | - Shan Nan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, and Key Laboratory of Aquaculture Disease Control, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China.
| |
Collapse
|
3
|
Kim JH, Kim K, Kim I, Seong S, Koh JT, Kim N. MCP-5 suppresses osteoclast differentiation through Ccr5 upregulation. J Cell Physiol 2024; 239:e31171. [PMID: 38214098 DOI: 10.1002/jcp.31171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 01/13/2024]
Abstract
Human monocyte chemoattractant protein-1 (MCP-1) in mice has two orthologs, MCP-1 and MCP-5. MCP-1, which is highly expressed in osteoclasts rather than in osteoclast precursor cells, is an important factor in osteoclast differentiation. However, the roles of MCP-5 in osteoclasts are completely unknown. In this study, contrary to MCP-1, MCP-5 was downregulated during receptor activator of nuclear factor kappa B ligand (RANKL)-induced osteoclast differentiation and was considered an inhibitory factor in osteoclast differentiation. The inhibitory role of MCP-5 in osteoclast differentiation was closely related to the increase in Ccr5 expression and the inhibition of IκB degradation by RANKL. Transgenic mice expressing MCP-5 controlled by Mx-1 promoter exhibited an increased bone mass because of a decrease in osteoclasts. This result strongly supported that MCP-5 negatively regulated osteoclast differentiation. MCP-5 also prevented severe bone loss caused by RANKL.
Collapse
Affiliation(s)
- Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Jeong-Tae Koh
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Hard-Tissue Biointerface Research Center, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
4
|
Sung EA, Park MH, Song S, Alanya H, Henegariu O, Liu J, Erson-Omay EZ, Sime PJ, Chae WJ. Thrombocyte-derived Dickkopf1 promotes macrophage polarization in the Bleomycin-induced lung injury model. Front Immunol 2023; 14:1247330. [PMID: 38162655 PMCID: PMC10757334 DOI: 10.3389/fimmu.2023.1247330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 11/20/2023] [Indexed: 01/03/2024] Open
Abstract
Immune responses are crucial to maintaining tissue homeostasis upon tissue injury. Upon various types of challenges, macrophages play a central role in regulating inflammation and tissue repair processes. While an immunomodulatory role of Wnt antagonist Dickkopf1 (DKK1) has been implicated, the role of Wnt antagonist DKK1 in regulating macrophage polarization in inflammation and the tissue repair process remains elusive. Here we found that DKK1 induces gene expression profiles to promote inflammation and tissue repair in macrophages. Importantly, DKK1 induced various genes, including inflammation and tissue repair, via JNK (c-jun N-terminal kinase) in macrophages. Furthermore, DKK1 potentiated IL-13-mediated macrophage polarization and activation. The co-inhibition of JNK and STAT6 markedly decreased gene expressions relevant to inflammation and fibrosis by DKK1 and IL-13. Interestingly, thrombocyte-specific deletion of DKK1 in mice reduced collagen deposition and decreased Arg1, CD206, HIF1α, and IL1β protein expressions in monocyte-derived alveolar macrophages in the acute sterile bleomycin (BLM)-induced lung injury model. These data suggested that thrombocytes communicate with macrophages via DKK1 to orchestrate inflammation and repair in this model. Taken together, our study demonstrates DKK1's role as an important regulatory ligand for macrophage polarization in the injury-induced inflammation and repair process in the lung.
Collapse
Affiliation(s)
- Eun-Ah Sung
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Min Hee Park
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - SuJeong Song
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Hasan Alanya
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
| | - Octavian Henegariu
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
| | - Jinze Liu
- Department of Biostatistics, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - E Zeynep Erson-Omay
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, United States
| | - Patricia J. Sime
- Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Wook-Jin Chae
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Phillips Oral Health Research Institute, Virginia Commonwealth University School of Dentistry, Richmond, VA, United States
| |
Collapse
|
5
|
Fernandes HB, de Oliveira IM, Postler TS, Lima SQ, Santos CAC, Oliveira MS, Leão FB, Ghosh S, Souza MC, Andrade W, Silva AM. Transcriptomic analysis reveals that RasGEF1b deletion alters basal and LPS-induced expression of genes involved in chemotaxis and cytokine responses in macrophages. Sci Rep 2023; 13:19614. [PMID: 37950057 PMCID: PMC10638313 DOI: 10.1038/s41598-023-47040-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023] Open
Abstract
Ras guanine nucleotide exchange factor member 1b (RasGEF1b) of the RasGEF/CDC25 domain-containing family is preferentially expressed by macrophages. However, information is lacking about its role in macrophage function. In this study, we generated mice with ubiquitous deletion of Rasgef1b and used RNA-seq-based transcriptomics to compare the global gene expression in wild-type and knock-out primary bone-marrow-derived macrophages under basal conditions and after lipopolysaccharide (LPS) treatment. Transcriptional filtering identified several genes with significantly different transcript levels between wild-type and knock-out macrophages. In total, 49 and 37 differentially expressed genes were identified at baseline and in LPS-activated macrophages, respectively. Distinct biological processes were significantly linked to down-regulated genes at the basal condition only, and largely included chemotaxis, response to cytokines, and positive regulation of GTPase activity. Importantly, validation by RT-qPCR revealed that the expression of genes identified as down-regulated after LPS stimulation was also decreased in the knock-out cells under basal conditions. We used a luciferase-based reporter assay to showcase the capability of RasGEF1b in activating the Serpinb2 promoter. Notably, knockdown of RasGEF1b in RAW264.7 macrophages resulted in impaired transcriptional activation of the Serpinb2 promoter, both in constitutive and LPS-stimulated conditions. This study provides a small collection of genes that shows relative expression changes effected by the absence of RasGEF1b in macrophages. Thus, we present the first evidence that RasGEF1b mediates the regulation of both steady-state and signal-dependent expression of genes and propose that this GEF plays a role in the maintenance of the basal transcriptional level in macrophages.
Collapse
Affiliation(s)
- Heliana B Fernandes
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Isadora Mafra de Oliveira
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
- Faculdade de Medicina de Ribeirão Preto, Av. Bandeirantes, 3900 - Campus da USP, Ribeirão Preto, SP, 14049-900, Brazil
| | - Thomas S Postler
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Design and Development Laboratory, International AIDS Vaccine Initiative, Brooklyn, NY, USA
| | - Sérgio Q Lima
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Cícera A C Santos
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
- Instituto Federal de Educação, Ciência e Tecnologia de Rondônia (IFRO), Guajará-Mirim, RO, Brazil
| | - Michaelle S Oliveira
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil
| | - Felipe B Leão
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Sankar Ghosh
- Department of Microbiology & Immunology, Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Maria C Souza
- Faculdade de Medicina de Ribeirão Preto, Av. Bandeirantes, 3900 - Campus da USP, Ribeirão Preto, SP, 14049-900, Brazil
| | - Warrison Andrade
- Faculdade de Medicina de Ribeirão Preto, Av. Bandeirantes, 3900 - Campus da USP, Ribeirão Preto, SP, 14049-900, Brazil
| | - Aristóbolo M Silva
- Laboratory of Inflammatory Genes, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
6
|
Azarfar F, Abbasi B, Jalali A, Abbasian MH. Investigation of the relationship between monocyte chemoattractant protein 1 rs1024611 variant and severity of COVID-19. Cytokine 2023; 171:156367. [PMID: 37713941 DOI: 10.1016/j.cyto.2023.156367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/17/2023]
Abstract
BACKGROUND Higher expression of Monocyte Chemoattractant Protein 1 (MCP-1) was reported in several studies. The clinical severity of Coronavirus disease 2019 (COVID-19) could be affected by genetic polymorphisms in MCP-1. This study aimed to examine the impact of MCP-1 2518A/G polymorphism and clinical parameters with COVID-19 severity. METHODS The polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method was used for MCP-1 rs1024611 (A/G) genotyping in 116 outpatients, hospitalized, and ICU patients. The biochemical and hematological profiles were collected from the patient's medical records. RESULTS Based on the statistical analysis, there was no significant relationship between the -2518A/G (rs1024611) genetic polymorphism in the regulatory region of the MCP-1 gene and the severity of the COVID-19. Multivariate logistic regression analysis has shown that the severity of COVID-19 infection was associated with decreased levels of eosinophils, neutrophils, lymphocytes, and, monocyte and higher levels of SGPT, SGOT, NLR, CRP, ferritin, urea, and D-Dimer (P < 0.05). CONCLUSION The MCP-1 gene polymorphism had no impact on COVID-19 severity. However, to confirm these results, a large-scale study needs to be conducted.
Collapse
Affiliation(s)
- Fatemeh Azarfar
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran
| | - Bahareh Abbasi
- Department of Medical Genetics, National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| | - Amir Jalali
- Department of Biology, Faculty of Sciences, Arak University, Arak, Iran.
| | - Mohammad Hadi Abbasian
- Department of Medical Genetics, National Institute for Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
7
|
Cooper ID, Kyriakidou Y, Edwards K, Petagine L, Seyfried TN, Duraj T, Soto-Mota A, Scarborough A, Jacome SL, Brookler K, Borgognoni V, Novaes V, Al-Faour R, Elliott BT. Ketosis Suppression and Ageing (KetoSAge): The Effects of Suppressing Ketosis in Long Term Keto-Adapted Non-Athletic Females. Int J Mol Sci 2023; 24:15621. [PMID: 37958602 PMCID: PMC10650498 DOI: 10.3390/ijms242115621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Most studies on ketosis have focused on short-term effects, male athletes, or weight loss. Hereby, we studied the effects of short-term ketosis suppression in healthy women on long-standing ketosis. Ten lean (BMI 20.5 ± 1.4), metabolically healthy, pre-menopausal women (age 32.3 ± 8.9) maintaining nutritional ketosis (NK) for > 1 year (3.9 years ± 2.3) underwent three 21-day phases: nutritional ketosis (NK; P1), suppressed ketosis (SuK; P2), and returned to NK (P3). Adherence to each phase was confirmed with daily capillary D-beta-hydroxybutyrate (BHB) tests (P1 = 1.9 ± 0.7; P2 = 0.1 ± 0.1; and P3 = 1.9 ± 0.6 pmol/L). Ageing biomarkers and anthropometrics were evaluated at the end of each phase. Ketosis suppression significantly increased: insulin, 1.78-fold from 33.60 (± 8.63) to 59.80 (± 14.69) pmol/L (p = 0.0002); IGF1, 1.83-fold from 149.30 (± 32.96) to 273.40 (± 85.66) µg/L (p = 0.0045); glucose, 1.17-fold from 78.6 (± 9.5) to 92.2 (± 10.6) mg/dL (p = 0.0088); respiratory quotient (RQ), 1.09-fold 0.66 (± 0.05) to 0.72 (± 0.06; p = 0.0427); and PAI-1, 13.34 (± 6.85) to 16.69 (± 6.26) ng/mL (p = 0.0428). VEGF, EGF, and monocyte chemotactic protein also significantly increased, indicating a pro-inflammatory shift. Sustained ketosis showed no adverse health effects, and may mitigate hyperinsulinemia without impairing metabolic flexibility in metabolically healthy women.
Collapse
Affiliation(s)
- Isabella D. Cooper
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Yvoni Kyriakidou
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Kurtis Edwards
- Cancer Biomarkers and Mechanisms Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK;
| | - Lucy Petagine
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Thomas N. Seyfried
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA; (T.N.S.); (T.D.)
| | - Tomas Duraj
- Biology Department, Boston College, Chestnut Hill, MA 02467, USA; (T.N.S.); (T.D.)
| | - Adrian Soto-Mota
- Metabolic Diseases Research Unit, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City 14080, Mexico;
- Tecnologico de Monterrey, School of Medicine, Mexico City 14380, Mexico
| | - Andrew Scarborough
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Sandra L. Jacome
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Kenneth Brookler
- Retired former Research Collaborator, Aerospace Medicine and Vestibular Research Laboratory, Mayo Clinic, Scottsdale, AZ 85259, USA;
| | - Valentina Borgognoni
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Vanusa Novaes
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Rima Al-Faour
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| | - Bradley T. Elliott
- Ageing Biology and Age-Related Diseases, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK; (Y.K.); (L.P.); (A.S.); (S.L.J.); (V.B.); (V.N.); (R.A.-F.); (B.T.E.)
| |
Collapse
|
8
|
Klein M, Dijoux E, Cheminant MA, Intes L, Bouchaud G. GliSODin® prevents airway inflammation by inhibiting T-cell differentiation and activation in a mouse model of asthma. FRONTIERS IN ALLERGY 2023; 4:1199355. [PMID: 37346413 PMCID: PMC10279865 DOI: 10.3389/falgy.2023.1199355] [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: 04/03/2023] [Accepted: 05/18/2023] [Indexed: 06/23/2023] Open
Abstract
Background Asthma is a chronic inflammatory airway disease characterized by a prevailing type 2 inflammation, airway hyperresponsiveness, and mucus hypersecretion and is driven by various factors among which oxidative molecules, called reactive oxygen species (ROS), play a major role. Superoxide dismutases (SODs) are enzymes that constitute the first line of defense against ROS. Melon SOD-gliadin, which is known as GliSODin®, is commonly used as a nutritional supplement that has proven antioxidant properties. Objectives In this study, we evaluated the efficacy and mechanism of action GliSODin® in the treatment of allergic asthma. Methods House dust mite (HDM)-induced asthmatic mice were orally exposed to GliSODin®, and airway hyperresponsiveness, lung inflammation, in vitro T-cell polarization, in vivo T-cell reactivation, and blood immunoglobulin were investigated. Results GliSODin® reduced airway hyperresponsiveness, lung innate and adaptive immune response, and HDM-specific IgE production. Coculturing CD4+ T-cell with HDM-sensitized dendritic cells and GliSODin® reduced T-cell polarization into Th2 and Th17 cells. Moreover, adoptively transferred CD4+ T cells from asthmatic mice exhibited a reduced reactivation of Th2 and Th17 cells following stimulation with HDM plus GliSODin®. Conclusion GliSODin® abrogates asthma features and reduces CD4+ T-cell polarization and reactivation. Taken together, these data suggest that GliSODin® could be used for the management of asthma symptoms.
Collapse
Affiliation(s)
- Martin Klein
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Département de Médecine, Université Laval, Québec, QC, Canada
| | - Eleonore Dijoux
- Université de Nantes, CNRS, INSERM, l’institut du thorax, Nantes, France
| | | | | | - Grégory Bouchaud
- INRAE, Biopolymères Intéractions Assemblages (BIA), Nantes, France
| |
Collapse
|
9
|
Bogacka J, Pawlik K, Ciapała K, Ciechanowska A, Mika J. CC Chemokine Receptor 4 (CCR4) as a Possible New Target for Therapy. Int J Mol Sci 2022; 23:ijms232415638. [PMID: 36555280 PMCID: PMC9779674 DOI: 10.3390/ijms232415638] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Chemokines and their receptors participate in many biological processes, including the modulation of neuroimmune interactions. Approximately fifty chemokines are distinguished in humans, which are classified into four subfamilies based on the N-terminal conserved cysteine motifs: CXC, CC, C, and CX3C. Chemokines activate specific receptors localized on the surface of various immune and nervous cells. Approximately twenty chemokine receptors have been identified, and each of these receptors is a seven-transmembrane G-protein coupled receptor. Recent studies provide new evidence that CC chemokine receptor 4 (CCR4) is important in the pathogenesis of many diseases, such as diabetes, multiple sclerosis, asthma, dermatitis, and cancer. This review briefly characterizes CCR4 and its ligands (CCL17, CCL22, and CCL2), and their contributions to immunological and neoplastic diseases. The review notes a significant role of CCR4 in nociceptive transmission, especially in painful neuropathy, which accompanies many diseases. The pharmacological blockade of CCR4 seems beneficial because of its pain-relieving effects and its influence on opioid efficacy. The possibilities of using the CCL2/CCL17/CCL22/CCR4 axis as a target in new therapies for many diseases are also discussed.
Collapse
Affiliation(s)
| | | | | | | | - Joanna Mika
- Correspondence: or ; Tel.: +48-12-6623-298; Fax: +48-12-6374-500
| |
Collapse
|
10
|
Ranjbar M, Rahimi A, Baghernejadan Z, Ghorbani A, Khorramdelazad H. Role of CCL2/CCR2 axis in the pathogenesis of COVID-19 and possible Treatments: All options on the Table. Int Immunopharmacol 2022; 113:109325. [PMID: 36252475 PMCID: PMC9561120 DOI: 10.1016/j.intimp.2022.109325] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/05/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is cause of the novel coronavirus disease (COVID-19). In the last two years, SARS-CoV-2 has infected millions of people worldwide with different waves, resulting in the death of many individuals. The evidence disclosed that the host immune responses to SARS-CoV-2 play a pivotal role in COVID-19 pathogenesis and clinical manifestations. In addition to inducing antiviral immune responses, SARS-CoV-2 can also cause dysregulated inflammatory responses characterized by the noticeable release of proinflammatory mediators in COVID-19 patients. Among these proinflammatory mediators, chemokines are considered a subset of cytokines that participate in the chemotaxis process to recruit immune and non-immune cells to the site of inflammation and infection. Researchers have demonstrated that monocyte chemoattractant protein-1 (MCP-1/CCL2) and its receptor (CCR2) are involved in the recruitment of monocytes and infiltration of these cells into the lungs of patients suffering from COVID-19. Moreover, elevated levels of CCL2 have been reported in the bronchoalveolar lavage fluid (BALF) obtained from patients with severe COVID-19, initiating cytokine storm and promoting CD163+ myeloid cells infiltration in the airways and further alveolar damage. Therefore, CCL2/CCR axis plays a key role in the immunopathogenesis of COVID-19 and targeted therapy of involved molecules in this axis can be a potential therapeutic approach for these patients. This review discusses the biology of the CCL2/CCR2 axis as well as the role of this axis in COVID-19 immunopathogenesis, along with therapeutic options aimed at inhibiting CCL2/CCR2 and modulating dysregulated inflammatory responses in patients with severe SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Mitra Ranjbar
- Department of Infectious Disease, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Atousa Ghorbani
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
11
|
Yi L, Zhou Y, Song J, Tang W, Yu H, Huang X, Shi H, Chen M, Sun J, Wei Y, Dong J. A novel iridoid glycoside leonuride (ajugol) attenuates airway inflammation and remodeling through inhibiting type-2 high cytokine/chemokine activity in OVA-induced asthmatic mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154345. [PMID: 35905568 DOI: 10.1016/j.phymed.2022.154345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/12/2022] [Accepted: 07/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Asthma is a chronic airway disorder with a hallmark feature of airflow obstruction that associated with the remodeling and inflammation in the airway wall. Effective therapy for controlling both remodeling and inflammation is still urgently needed. Leonuride is the main pharmacological component identified from Bu-Shen-Yi-Qi-Tang (BSYQT) which has been traditionally used in treatment of lung diseases. However, no pharmacological effects of leonuride in asthma were reported. PURPOSE Here we aimed to investigated whether leonuride provided a therapeutic efficacy in reversing asthma airway remodeling and inflammation and uncover the underlying mechanisms. STUDY DESIGN AND METHODS Mouse models of chronic asthma were developed with ovalbumin (OVA) exposure for 8 weeks. Respiratory mechanics, lung histopathology and asthma-related cytokines were examined. Lung tissues were analyzed using RNA sequencing to reveal the transcriptional profiling changes. RESULTS After oral administration with leonuride (15 mg/kg or 30 mg/kg), mice exhibited a lower airway hyperresponsiveness in comparison to asthmatic mice. Leonuride suppressed airway inflammation evidenced by the significant reductions in accumulation of inflammatory cells around bronchi and vessels, leukocyte population counts and the abundance of type 2 inflammatory mediators (OVA specific IgE, IL-4, IL-5 and IL-13) in bronchoalveolar lavage fluid (BALF). On the other hand, leonuride slowed down the process of active remodeling as demonstrated by weaker goblet cell metaplasia and subepithelial fibrosis in lung histopathology and lower transforming growth factor (TGF)-β1 levels in serum and BALF in comparison to mice treated with OVA only. Furthermore, we uncovered transcriptional profiling alternations in lung tissue of mice after OVA exposure and leonuride treatment. Gene sets belonging to type-2 cytokine/chemokine activity stood out in leonuride target transcripts. Those upregulated (Bmp10, Ccl12, Ccl22, Ccl8, Ccl9, Cxcl15, Il13, Il33, Tnfrsf9, Il31ra, Il5ra, Il13ra2 and Ccl24) or downregulated (Acvr1c and Il18) genes in asthmatic mice, were all reversely regulated by leonuride treatment. CONCLUSIONS Our results revealed the therapeutic efficacy of leonuride in experimental chronic asthma for the first time, and implied that its anti-inflammatory and antifibrotic properties might be mediated by regulation of type-2 high cytokine/chemokines responses.
Collapse
Affiliation(s)
- La Yi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yaolong Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingrong Song
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Hanlin Shi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Mengmeng Chen
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jing Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China.
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China; Institutes of Integrative Medicine, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Bantulà M, Tubita V, Roca-Ferrer J, Mullol J, Valero A, Bobolea I, Pascal M, de Hollanda A, Vidal J, Picado C, Arismendi E. Differences in Inflammatory Cytokine Profile in Obesity-Associated Asthma: Effects of Weight Loss. J Clin Med 2022; 11:jcm11133782. [PMID: 35807067 PMCID: PMC9267201 DOI: 10.3390/jcm11133782] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023] Open
Abstract
Obesity and asthma are associated with systemic inflammation maintained by mediators released by adipose tissue and lung. This study investigated the inflammatory serum mediator profile in obese subjects (O) (n = 35), non-obese asthma (NOA) patients (n = 14), obese asthmatics (OA) (n = 21) and healthy controls (HC) (n = 33). The effect of weight loss after bariatric surgery (BS) was examined in 10 OA and 31 O subjects. We analyzed serum markers including leptin, adiponectin, TGF-β1, TNFR2, MCP-1, ezrin, YKL-40, ST2, IL-5, IL-9, and IL-18. Compared with HC subjects, the O group showed increased levels of leptin, TGF-β1, TNFR2, MCP-1, ezrin, YKL-40, and ST2; the OA group presented increased levels of MCP-1, ezrin, YKL-40, and IL-18, and the NOA group had increased levels of ezrin, YKL-40, IL-5, and IL-18. The higher adiponectin/leptin ratio in NOA with respect to OA subjects was the only significant difference between the two groups. IL-9 was the only cytokine with significantly higher levels in OA with respect to O subjects. TNFR2, ezrin, MCP-1, and IL-18 concentrations significantly decreased in O subjects after BS. O, OA, and NOA showed distinct patterns of systemic inflammation. Leptin and adiponectin are regulated in asthma by obesity-dependent and -independent mechanisms. Combination of asthma and obesity does not result in significant additive effects on circulating cytokine levels.
Collapse
Affiliation(s)
- Marina Bantulà
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
- Correspondence: ; Tel.: +34-932275400
| | - Valeria Tubita
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
| | - Jordi Roca-Ferrer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Joaquim Mullol
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Rhinology Unit & Smell Clinic, ENT Department, Hospital Clinic, 08036 Barcelona, Spain
| | - Antonio Valero
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Pulmonology and Allergy Department, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Irina Bobolea
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Pulmonology and Allergy Department, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Mariona Pascal
- Immunology Department, CDB, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain;
| | - Ana de Hollanda
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic, 08036 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Fisopatología de la Obesidad y Nutrición (CIBEROBN), 28029 Madrid, Spain
| | - Josep Vidal
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
- Obesity Unit, Endocrinology and Nutrition Department, Hospital Clínic, 08036 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red en Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
| | - César Picado
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Pulmonology and Allergy Department, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| | - Ebymar Arismendi
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (V.T.); (J.R.-F.); (J.M.); (A.V.); (I.B.); (A.d.H.); (J.V.); (C.P.); (E.A.)
- Centro de Investigaciones Biomédicas en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Pulmonology and Allergy Department, Hospital Clinic, University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
13
|
Shaghayegh G, Cooksley C, Ramezanpour M, Wormald PJ, Psaltis AJ, Vreugde S. Chronic Rhinosinusitis, S. aureus Biofilm and Secreted Products, Inflammatory Responses, and Disease Severity. Biomedicines 2022; 10:1362. [PMID: 35740385 PMCID: PMC9220248 DOI: 10.3390/biomedicines10061362] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic rhinosinusitis (CRS) is a persistent inflammation of the nasal cavity and paranasal sinuses associated with tissue remodelling, dysfunction of the sinuses' natural defence mechanisms, and induction of different inflammatory clusters. The etiopathogenesis of CRS remains elusive, and both environmental factors, such as bacterial biofilms and the host's general condition, are thought to play a role. Bacterial biofilms have significant clinical relevance due to their potential to cause resistance to antimicrobial therapy and host defenses. Despite substantial medical advances, some CRS patients suffer from recalcitrant disease that is unresponsive to medical and surgical treatments. Those patients often have nasal polyps with tissue eosinophilia, S. aureus-dominant mucosal biofilm, comorbid asthma, and a severely compromised quality of life. This review aims to summarise the contemporary knowledge of inflammatory cells/pathways in CRS, the role of bacterial biofilm, and their impact on the severity of the disease. Here, an emphasis is placed on S. aureus biofilm and its secreted products. A better understanding of these factors might offer important diagnostic and therapeutic perceptions for recalcitrant disease.
Collapse
Affiliation(s)
- Gohar Shaghayegh
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Clare Cooksley
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Mahnaz Ramezanpour
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Peter-John Wormald
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Alkis James Psaltis
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| | - Sarah Vreugde
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5000, Australia; (G.S.); (C.C.); (M.R.); (P.-J.W.); (A.J.P.)
- Department of Surgery-Otolaryngology-Head and Neck Surgery, University of Adelaide, Adelaide 5011, Australia
- Central Adelaide Local Health Network, The Queen Elizabeth Hospital, The Basil Hetzel Institute for Translational Health Research, Woodville South 5011, Australia
| |
Collapse
|
14
|
Fu Y, Wang J, Zhou B, Pajulas A, Gao H, Ramdas B, Koh B, Ulrich BJ, Yang S, Kapur R, Renauld JC, Paczesny S, Liu Y, Tighe RM, Licona-Limón P, Flavell RA, Takatsuka S, Kitamura D, Tepper RS, Sun J, Kaplan MH. An IL-9-pulmonary macrophage axis defines the allergic lung inflammatory environment. Sci Immunol 2022; 7:eabi9768. [PMID: 35179949 PMCID: PMC8991419 DOI: 10.1126/sciimmunol.abi9768] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite IL-9 functioning as a pleiotropic cytokine in mucosal environments, the IL-9-responsive cell repertoire is still not well defined. Here, we found that IL-9 mediates proallergic activities in the lungs by targeting lung macrophages. IL-9 inhibits alveolar macrophage expansion and promotes recruitment of monocytes that develop into CD11c+ and CD11c- interstitial macrophage populations. Interstitial macrophages were required for IL-9-dependent allergic responses. Mechanistically, IL-9 affected the function of lung macrophages by inducing Arg1 activity. Compared with Arg1-deficient lung macrophages, Arg1-expressing macrophages expressed greater amounts of CCL5. Adoptive transfer of Arg1+ lung macrophages but not Arg1- lung macrophages promoted allergic inflammation that Il9r-/- mice were protected against. In parallel, the elevated expression of IL-9, IL-9R, Arg1, and CCL5 was correlated with disease in patients with asthma. Thus, our study uncovers an IL-9/macrophage/Arg1 axis as a potential therapeutic target for allergic airway inflammation.
Collapse
Affiliation(s)
- Yongyao Fu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jocelyn Wang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Baohua Zhou
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Abigail Pajulas
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Baskar Ramdas
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Byunghee Koh
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Benjamin J Ulrich
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shuangshuang Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Reuben Kapur
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jean-Christophe Renauld
- Ludwig Institute for Cancer Research, Experimental Medicine Unit, Université Catholique de Louvain, Brussels, 1200 Belgium
| | - Sophie Paczesny
- Department of Microbiology and Immunology, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Robert M Tighe
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Paula Licona-Limón
- Departamento de Biologia Celular y del Desarrollo, Instituto de Fisiologia Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shogo Takatsuka
- Division of Molecular Biology, Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, Noda, Japan
| | - Daisuke Kitamura
- Division of Molecular Biology, Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, Noda, Japan
| | - Robert S. Tepper
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jie Sun
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
15
|
Racca F, Pellegatta G, Cataldo G, Vespa E, Carlani E, Pelaia C, Paoletti G, Messina MR, Nappi E, Canonica GW, Repici A, Heffler E. Type 2 Inflammation in Eosinophilic Esophagitis: From Pathophysiology to Therapeutic Targets. Front Physiol 2022; 12:815842. [PMID: 35095572 PMCID: PMC8790151 DOI: 10.3389/fphys.2021.815842] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022] Open
Abstract
Eosinophilic esophagitis (EoE) is a chronic immune-mediated disease of the esophagus characterized clinically by symptoms related to esophageal dysfunction and histologically by eosinophil-predominant inflammation, whose incidence is rising. It significantly affects patients’ quality of life and, if left untreated, results in fibrotic complications. Although broad consensus has been achieved on first-line therapy, a subset of patients remains non-responder to standard therapy. The pathogenesis of EoE is multifactorial and results from the complex, still mostly undefined, interaction between genetics and intrinsic factors, environment, and antigenic stimuli. A deep understanding of the pathophysiology of this disease is pivotal for the development of new therapies. This review provides a comprehensive description of the pathophysiology of EoE, starting from major pathogenic mechanisms (genetics, type 2 inflammation, epithelial barrier dysfunction, gastroesophageal reflux, allergens, infections and microbiota) and subsequently focusing on the single protagonists of type 2 inflammation (involved cells, cytokines, soluble effectors, surface proteins and transcription factors) that could represent present and future therapeutic targets, while summarizing previous therapeutic approaches in literature.
Collapse
Affiliation(s)
- Francesca Racca
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- *Correspondence: Francesca Racca,
| | - Gaia Pellegatta
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Giuseppe Cataldo
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Edoardo Vespa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Elisa Carlani
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Corrado Pelaia
- Department of Medical and Surgical Sciences, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Giovanni Paoletti
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Maria Rita Messina
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Emanuele Nappi
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Giorgio Walter Canonica
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Alessandro Repici
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Italy
| | - Enrico Heffler
- Personalized Medicine, Asthma and Allergy, IRCCS Humanitas Research Hospital, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| |
Collapse
|
16
|
Ishii T, Murakami Y, Narita T, Nunokawa H, Miyake K, Nagase T, Yamashita N. Myeloid differentiation protein-2 has a protective role in house dust mite-mediated asthmatic characteristics with the proinflammatory regulation of airway epithelial cells and dendritic cells. Clin Exp Allergy 2021; 52:149-161. [PMID: 34418187 DOI: 10.1111/cea.14002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 08/17/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Myeloid differentiation protein-2 (MD-2) is a lipopolysaccharide-binding protein involved in lipopolysaccharide signalling via Toll-like receptor 4 (TLR4). TLR4 plays an essential role in HDM-mediated allergic airway inflammation. Moreover, MD-2 is structurally similar to Der f 2, a major allergen from house dust mite (HDM). OBJECTIVES We aimed to clarify the role of MD-2 in the pathogenesis of HDM-mediated allergic airway inflammation. METHODS Wild-type (WT), TLR4 knockout and MD-2 knockout mice were subjected to intranasal instillation of HDM extract, and asthmatic features were evaluated. We also evaluated gene sets regulated by MD-2 in HDM-treated airway epithelial cells and examined the function of dendritic cells from lymph nodes and from lungs. RESULTS Aggravated allergic airway inflammation with increased airway hyperresponsiveness was observed in MD-2 knockout mice compared with WT and TLR4 knockout mice. Global gene expression analysis revealed an MD-2 regulated proinflammatory response and reconstituted TLR4 signalling in airway epithelial cells. The ability of dendritic cells to evoke an allergic immune response was enhanced in MD-2 knockout mice. CONCLUSIONS & CLINICAL RELEVANCE MD-2 plays a protective role in HDM-induced airway allergy with the proinflammatory regulation of airway epithelial cells and dendritic cells. MD-2 may serve as a therapeutic target in the treatment of asthma.
Collapse
Affiliation(s)
- Takashi Ishii
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan.,Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Murakami
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| | - Tomoya Narita
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| | - Hiroki Nunokawa
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takahide Nagase
- Department of Respiratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naomi Yamashita
- Department of Pharmacotherapy, Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| |
Collapse
|
17
|
Cultured human mast cells release various chemokines after stimulation with IL-33. Allergol Int 2021; 70:386-388. [PMID: 33583698 DOI: 10.1016/j.alit.2020.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 12/22/2020] [Accepted: 12/27/2020] [Indexed: 11/20/2022] Open
|
18
|
Branchett WJ, Cook J, Oliver RA, Bruno N, Walker SA, Stölting H, Mack M, O'Garra A, Saglani S, Lloyd CM. Airway macrophage-intrinsic TGF-β1 regulates pulmonary immunity during early-life allergen exposure. J Allergy Clin Immunol 2021; 147:1892-1906. [PMID: 33571538 PMCID: PMC8098862 DOI: 10.1016/j.jaci.2021.01.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Early life represents a major risk window for asthma development. However, the mechanisms controlling the threshold for establishment of allergic airway inflammation in early life are incompletely understood. Airway macrophages (AMs) regulate pulmonary allergic responses and undergo TGF-β-dependent postnatal development, but the role of AM maturation factors such as TGF-β in controlling the threshold for pathogenic immune responses to inhaled allergens remains unclear. OBJECTIVE Our aim was to test the hypothesis that AM-derived TGF-β1 regulates pathogenic immunity to inhaled allergen in early life. METHODS Conditional knockout (Tgfb1ΔCD11c) mice, with TGF-β1 deficiency in AMs and other CD11c+ cells, were analyzed throughout early life and following neonatal house dust mite (HDM) inhalation. The roles of specific chemokine receptors were determined by using in vivo blocking antibodies. RESULTS AM-intrinsic TGF-β1 was redundant for initial population of the neonatal lung with AMs, but AMs from Tgfb1ΔCD11c mice failed to adopt a mature homeostatic AM phenotype in the first weeks of life. Evidence of constitutive TGF-β1 signaling was also observed in pediatric human AMs. TGF-β1-deficient AMs expressed enhanced levels of monocyte-attractant chemokines, and accordingly, Tgfb1ΔCD11c mice exposed to HDM throughout early life accumulated CCR2-dependent inflammatory CD11c+ mononuclear phagocytes into the airway niche that expressed the proallergic chemokine CCL8. Tgfb1ΔCD11c mice displayed augmented TH2, group 2 innate lymphoid cell, and airway remodeling responses to HDM, which were ameliorated by blockade of the CCL8 receptor CCR8. CONCLUSION Our results highlight a causal relationship between AM maturity, chemokines, and pathogenic immunity to environmental stimuli in early life and identify TGF-β1 as a key regulator of this.
Collapse
Affiliation(s)
- William J Branchett
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - James Cook
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, United Kingdom
| | - Robert A Oliver
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Nicoletta Bruno
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Simone A Walker
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Helen Stölting
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Matthias Mack
- Department of Internal Medicine II- Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Anne O'Garra
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom; Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London, United Kingdom
| | - Sejal Saglani
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom; Department of Paediatric Respiratory Medicine, Royal Brompton Hospital, London, United Kingdom
| | - Clare M Lloyd
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom.
| |
Collapse
|
19
|
Darling NJ, Arthur JSC, Cohen P. Salt-inducible kinases are required for the IL-33-dependent secretion of cytokines and chemokines in mast cells. J Biol Chem 2021; 296:100428. [PMID: 33600797 PMCID: PMC7988334 DOI: 10.1016/j.jbc.2021.100428] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/28/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023] Open
Abstract
Cytokines and chemokines are important regulators of airway hyper-responsiveness, immune cell infiltration, and inflammation and are produced when mast cells are stimulated with interleukin-33 (IL-33). Here, we establish that the salt-inducible kinases (SIKs) are required for the IL-33-stimulated transcription of il13, gm-csf and tnf and hence the production of these cytokines. The IL-33-stimulated secretion of IL-13, granulocyte-macrophage colony stimulating factor, and tumor necrosis factor was strongly reduced in fetal liver-derived mast cells from mice expressing a kinase-inactive mutant of SIK3 and abolished in cells expressing kinase-inactive mutants of SIK2 and SIK3. The IL-33-dependent secretion of these cytokines and several chemokines was also abolished in SIK2/3 double knock-out bone marrow-derived mast cells (BMMC), reduced in SIK3 KO cells but little affected in BMMC expressing kinase-inactive mutants of SIK1 and SIK2 or lacking SIK2 expression. In SIK2 knock-out BMMC, the expression of SIK3 was greatly increased. Our studies identify essential roles for SIK2 and SIK3 in producing inflammatory mediators that trigger airway inflammation. The effects of SIKs were independent of IκB kinase β, IκB kinase β-mediated NF-κB-dependent gene transcription, and activation of the mitogen-activated protein kinase family members p38α and c-jun N-terminal kinases. Our results suggest that dual inhibitors of SIK2 and SIK3 may have therapeutic potential for the treatment of mast cell-driven diseases.
Collapse
Affiliation(s)
- Nicola J Darling
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Angus, UK
| | - J Simon C Arthur
- Division of Cell Signalling and Immunology, University of Dundee, Dundee, Angus, UK
| | - Philip Cohen
- MRC Protein Phosphorylation and Ubiquitylation Unit, University of Dundee, Dundee, Angus, UK.
| |
Collapse
|
20
|
Jiang L, Li Y, Wang F, Zhang X, Zhao R. Protective Effect of S-Allyl Cysteine Against Neonatal Asthmatic Rats. Dose Response 2020; 18:1559325820982189. [PMID: 33488313 PMCID: PMC7768841 DOI: 10.1177/1559325820982189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 11/22/2022] Open
Abstract
S-Allyl cysteine (SAC), an organic compound and a natural constituent of Allium sativum, commonly known as garlic have been consumed in routine foods are known to possess various biological activities. Nevertheless, scientific evidence on the protective effect of SAC against neonatal asthmatic rats is not available. Hence, the present study aimed at investigating the anti-asthmatic activity of SAC in neonatal asthmatic rats using Wistar rats. The study conducted in 4 groups consists of normal control rats, asthma-induced, asthma animals administered with SAC (25 mg/kg), and SAC control. At the end of the experimental period, inflammatory cells in bronchoalveolar lavage fluid (BALF), inflammatory markers, fibrinogen level, activated partial thromboplastin time, coagulation factor activity, and histopathology were elucidated. The current investigation exhibits that SAC significantly reduced the total leukocytes, with restored fibrinogen level, and activated partial thromboplastin time. In addition, the levels of inflammatory cytokines such as TNF-α (tumor necrosis factor- α), IL-6 (Interleukin 6), and IL-1β have also attenuated in SAC treated animals. Furthermore, the mRNA expression levels of COX2 (cyclooxygenase-2), MCP-1 (monocyte chemoattractant protein-1), RANTES (regulated upon activation, normal T cell expressed and secreted), and eotaxin were reduced in SAC treated animals. Treatment of rats with SAC significantly reduced inflammation and eosinophil infiltration in the lungs. These results suggest that SAC exert protection in neonatal asthmatic rats suffering from acute or chronic inflammation by inducing anti-inflammatory and cell-protective responses.
Collapse
Affiliation(s)
- Li Jiang
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuning Li
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Fang Wang
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xindao Zhang
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ruiping Zhao
- Department of Pediatrics, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
21
|
Yanagisawa R, Koike E, Win-Shwe TT, Kawaguchi M, Takano H. The impact of oral exposure to low-dose tris(2-butoxyethyl) phosphate in allergic asthmatic mice. J Appl Toxicol 2020; 40:1498-1510. [PMID: 32497324 DOI: 10.1002/jat.4001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 12/24/2022]
Abstract
Tris(2-butoxyethyl) phosphate (TBEP) is a major organophosphorus flame retardant and has been widely increasing as a substitute for brominated flame retardants. TBEP may have adverse effects on human health; however, its impact on immune and allergic responses remains largely uncharacterized. In this study, the effects of low-dose TBEP comparable with the level of actual human exposure to that of human tolerable daily intake on allergic asthmatic mice were explored. Five-week-old C3H/HeJSlc male mice consumed a diet containing approximately 0.02, 0.2 or 2 μg/kg/day TBEP and were intratracheally administrated ovalbumin (OVA) (1 μg/mouse every 2 weeks from 5 to 11 weeks of age). Exposure to 2 μg/kg/day TBEP with OVA tended to enhance allergic pulmonary inflammation and significantly elevated mRNA levels of interleukin-5, eotaxin-1 and estrogen receptor alpha (ERα) compared with OVA alone. In mediastinal lymph nodes (MLNs), TBEP (0.2 or 2 μg/kg/day) with OVA significantly increased in total cell number and promoted conventional dendritic cell activation than OVA alone; MLN cell proliferation by OVA restimulation was also enhanced in these groups. In the bone marrow (BM), TBEP (0.02 or 0.2 μg/kg/day) with OVA resulted in a net decrease in total cell number and fraction of CCR2+ Gr-1+ cells; the fraction of Gr-1+ cells increased. In conclusion, oral exposure to low-dose TBEP levels equivalent to tolerable daily intake may exacerbate allergic pulmonary inflammation by promoting a skewed T-helper 2 cell response, upregulation of ERα and dysregulation of both MLN and BM microenvironments.
Collapse
Affiliation(s)
- Rie Yanagisawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
| | - Eiko Koike
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
| | - Tin-Tin Win-Shwe
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Japan
| | | | - Hirohisa Takano
- Graduate School of Global Environmental Studies, Kyoto, Japan
| |
Collapse
|
22
|
Simon SCS, Hu X, Panten J, Grees M, Renders S, Thomas D, Weber R, Schulze TJ, Utikal J, Umansky V. Eosinophil accumulation predicts response to melanoma treatment with immune checkpoint inhibitors. Oncoimmunology 2020; 9:1727116. [PMID: 32117594 PMCID: PMC7028332 DOI: 10.1080/2162402x.2020.1727116] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 12/04/2019] [Accepted: 12/06/2019] [Indexed: 01/03/2023] Open
Abstract
Eosinophils have been identified as a prognostic marker in immunotherapy of melanoma and suggested to contribute to anti-tumor host defense. However, the influence of immune checkpoint inhibitors (ICI) on the eosinophil population is poorly studied. Here, we applied routine laboratory tests, multicolor flow cytometry, RNA microarray analysis, and bio-plex assay to analyze circulating eosinophils and related serum inflammatory factors in 32 patients treated with pembrolizumab or the combination of nivolumab and ipilimumab. We demonstrated that clinical responses to ICI treatment were associated with an eosinophil accumulation in the peripheral blood. Moreover, immunotherapy led to the alteration of the eosinophil genetic and activation profile. Elevated serum concentrations of IL-16 during ICI treatment were found to be associated with increased frequencies of eosinophils in the peripheral blood. Using immunohistochemistry, we observed an enhanced eosinophil degranulation and a positive correlation between eosinophil and CD8+ T cell infiltration of tumor tissues from melanoma patients treated with ICI. Our findings highlight additional mechanisms of ICI effects and suggest the level of eosinophils as a novel predictive marker for melanoma patients who may benefit from this immunotherapy.
Collapse
Affiliation(s)
- Sonja C S Simon
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Xiaoying Hu
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jasper Panten
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Research Group Hematopoietic and Leukemic Stem Cells, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Mareike Grees
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Simon Renders
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Research Group Hematopoietic and Leukemic Stem Cells, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Daniel Thomas
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rebekka Weber
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany.,Faculty of Biosciences, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | - Torsten J Schulze
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Diagnostics, Institute Springe, Springe, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
23
|
Chen W, Cui J, Xiang G, Zhang J, Gao H. Association between MCP-1 -2518A>G polymorphism and asthma susceptibility: a meta-analysis. ACTA ACUST UNITED AC 2019; 52:e8549. [PMID: 31664304 PMCID: PMC6826895 DOI: 10.1590/1414-431x20198549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 08/21/2019] [Indexed: 11/22/2022]
Abstract
The published data on the association between MCP-1 -2518A>G polymorphism and asthma susceptibility are inconclusive. Therefore, we performed a meta-analysis to estimate the impact of MCP-1 -2518A>G polymorphism on asthma susceptibility. PubMed, Web of Science, Wanfang, and China National Knowledge Infrastructure (CNKI) databases were used to identify eligible studies. The pooled odds ratios (ORs) and corresponding 95% confidence intervals (CIs) were used to calculate the strength of association. Sensitivity analysis was performed to evaluate the influence of individual studies on the estimates of overall effect, and funnel plots and Egger's test were used to assess publication bias. Eight publications with 1562 asthma patients and 1574 controls were finally identified. Overall, we found no significant association between MCP-1 -2518A>G polymorphism and asthma susceptibility in any of the genetic model comparisons. After stratified analysis by ethnicity, the results showed that a significant association with asthma risk was found in Caucasians in all the genetic models. However, a protective association was found in Africans under the dominant model. The present meta-analysis suggested that the MCP-1 -2518 A>G polymorphism is a risk factor for asthma in the Caucasian population, nevertheless it has a protective effect in the African population.
Collapse
Affiliation(s)
- Wenli Chen
- Respiratory Department, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jiewei Cui
- Respiratory Department, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Guoan Xiang
- Respiratory Department, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jianpeng Zhang
- Respiratory Department, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hongmei Gao
- Respiratory Department, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
24
|
Ayakannu R, Abdullah NA, Radhakrishnan AK, Lechimi Raj V, Liam CK. Relationship between various cytokines implicated in asthma. Hum Immunol 2019; 80:755-763. [PMID: 31054782 DOI: 10.1016/j.humimm.2019.04.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/18/2019] [Accepted: 04/26/2019] [Indexed: 01/01/2023]
Abstract
Asthma is a complex disorder involving immunologic, environmental, genetic and other factors. Today, asthma is the most common disease encountered in clinical medicine in both children and adults worldwide. Asthma is characterized by increased responsiveness of the tracheobronchial tree resulting in chronic swelling and inflammation of the airways recognized to be controlled by the T-helper 2 (Th2) lymphocytes, which secrete cytokines to increase the production of IgE by B cells. There are many cytokines implicated in the development of the chronic inflammatory processes that are often observed in asthma. Ultimately, these cytokines cause the release of mediators such as histamine and leukotrienes (LT), which in turn promote airway remodeling, bronchial hyperresponsiveness and bronchoconstriction. The CD4+ T-lymphocytes from the airways of asthmatics express a panel of cytokines that represent the Th2 cells. The knowledge derived from numerous experimental and clinical studies have allowed physicians and scientists to understand the normal functions of these cytokines and their roles in the pathogenesis of asthma. The main focus of this review is to accentuate the relationship between various cytokines implicated in human asthma. However, some key findings from animal models will be highlighted to support the discoveries from clinical studies.
Collapse
Affiliation(s)
- Rathimalar Ayakannu
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia; Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - N A Abdullah
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.
| | - Ammu K Radhakrishnan
- Jeffrey Cheah School of Medicine, Monash University Malaysia, Jalan Lagoon, 47500 Bandar Sunway, Selangor, Malaysia
| | - Vijaya Lechimi Raj
- Department of Pharmacology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Selangor, Malaysia
| | - C K Liam
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
25
|
Acetylcholine-treated murine dendritic cells promote inflammatory lung injury. PLoS One 2019; 14:e0212911. [PMID: 30822345 PMCID: PMC6396899 DOI: 10.1371/journal.pone.0212911] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 01/30/2019] [Indexed: 01/01/2023] Open
Abstract
In recent years a non-neuronal cholinergic system has been described in immune cells, which is often usually activated during the course of inflammatory processes. To date, it is known that Acetylcholine (ACh), a neurotransmitter extensively expressed in the airways, not only induces bronchoconstriction, but also promotes a set of changes usually associated with the induction of allergic/Th2 responses. We have previously demonstrated that ACh polarizes human dendritic cells (DC) toward a Th2-promoting profile through the activation of muscarinic acetylcholine receptors (mAChR). Here, we showed that ACh promotes the acquisition of an inflammatory profile by murine DC, with the increased MHC II IAd expression and production of two cytokines strongly associated with inflammatory infiltrate and tissue damage, namely TNF-α and MCP-1, which was prevented by blocking mAChR. Moreover, we showed that ACh induces the up-regulation of M3 mAChR expression and the blocking of this receptor with tiotropium bromide prevents the increase of MHC II IAd expression and TNF-α production induced by ACh on DC, suggesting that M3 is the main receptor involved in ACh-induced activation of DC. Then, using a short-term experimental murine model of ovalbumin-induced lung inflammation, we revealed that the intranasal administration of ACh-treated DC, at early stages of the inflammatory response, might be able to exacerbate the recruitment of inflammatory mononuclear cells, promoting profound structural changes in the lung parenchyma characteristic of chronic inflammation and evidenced by elevated systemic levels of inflammatory marker, TNF-α. These results suggest a potential role for ACh in the modulation of immune mechanisms underlying pulmonary inflammatory processes.
Collapse
|
26
|
Kubo F, Ariestanti DM, Oki S, Fukuzawa T, Demizu R, Sato T, Sabirin RM, Hirose S, Nakamura N. Loss of the adhesion G-protein coupled receptor ADGRF5 in mice induces airway inflammation and the expression of CCL2 in lung endothelial cells. Respir Res 2019; 20:11. [PMID: 30654796 PMCID: PMC6337809 DOI: 10.1186/s12931-019-0973-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 01/02/2019] [Indexed: 01/09/2023] Open
Abstract
Background Adhesion G-protein coupled receptor F5 (ADGRF5) was recently identified as an essential regulator of pulmonary surfactant homeostasis in alveolar type II cells. We previously showed that in addition to abnormal surfactant accumulation, Adgrf5-deficient (Adgrf5−/−) mice exhibit emphysema-like signs, suggesting a possible role for ADGRF5 in immune regulation. Here, we extended the phenotypic analysis of Adgrf5−/− mice to help understand its biological role in the lung, and especially in immune regulation. Methods Histological features of lungs were evaluated by Alcian blue and Masson’s trichrome staining. Quantitative real-time PCR (qPCR) and western blot analyses were performed to analyze the differential expression of genes/proteins related to airway inflammation in lungs between wildtype and Adgrf5−/− mice. Acid–base status was assessed by performing blood gas tests and urine pH measurements. Inflammatory cell counting was performed using Giemsa-stained bronchoalveolar lavage cells. Serum IgE concentrations were determined by enzyme-linked immunosorbent assay. The expression of Ccl2, S100a8, S100a9, and Saa3 in primary lung endothelial cells (ECs) was determined by qPCR and/or western blotting. Finally, the effect of administrating RS504393 to 2-week-old Adgrf5−/− mice on gene expression in the lungs was analyzed by qPCR. Results Adgrf5−/− mice exhibited several features of chronic airway inflammation (mucous cell metaplasia, mucus hyperproduction, subepithelial fibrosis, respiratory acidosis, high serum IgE, mast cell accumulation, and neutrophilia) in parallel with elevated expression of genes involved in mucous cell metaplasia (Muc5ac, Muc5b, Slc26a4, and Clca1), fibrosis (Tgfb1, Col1a1, Fn1, and Tnc), and type 2 immune response (Il4, Il5, Il13, IL-25, and IL-33) at 12 and/or 30 weeks of age. In contrast, mRNA expression of Ccl2, S100a8, and S100a9 was upregulated in embryonic or neonatal Adgrf5−/− lungs as well as in lung ECs of Adgrf5−/− mice at 1 week of age. RS504393 treatment suppressed the upregulation of S100a8, S100a9, Slc26a4, and Il5 in Adgrf5−/− lungs. Conclusions Targeted disruption of ADGRF5 results in the development of airway inflammation, which is likely mediated by the type 2 immune response and possibly CCL2-mediated inflammation. ADGRF5 also has a potential role in the regulation of genes encoding CCL2 in lung ECs, thereby maintaining immune homeostasis. Electronic supplementary material The online version of this article (10.1186/s12931-019-0973-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fumimasa Kubo
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Donna Maretta Ariestanti
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Souta Oki
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Taku Fukuzawa
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Ryotaro Demizu
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Tomoya Sato
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Rahmaningsih Mara Sabirin
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan.,Department of Physiology, Faculty of Medicine, Public Health and Nursing, Gadjah Mada University, JI.Farmako Sekip Utara, Yogyakarta, 55281, Indonesia
| | - Shigehisa Hirose
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan
| | - Nobuhiro Nakamura
- Department of Life Science and Technology, Tokyo Institute of Technology, 4259-B13 Nagatsuta-cho, Midori-ku, Yokohama, 226-8501, Japan.
| |
Collapse
|
27
|
Zhang K, Luo J. Role of MCP-1 and CCR2 in alcohol neurotoxicity. Pharmacol Res 2019; 139:360-366. [PMID: 30472461 PMCID: PMC6360095 DOI: 10.1016/j.phrs.2018.11.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/22/2018] [Accepted: 11/21/2018] [Indexed: 01/08/2023]
Abstract
Alcohol abuse causes profound damage to both the developing brain and the adult brain. Prenatal exposure to alcohol results in a wide range of deficits known as fetal alcohol spectrum disorders (FASD). Alcohol abuse in adults is associated with brain shrinkage, memory and attention deficits, communication disorders and physical disabilities. Monocyte chemoattractant protein-1 (MCP-1/CCL2) is one of the key chemokines that regulate the recruitment and activation of monocytes and microglia. Both MCP-1 and its receptor C-C chemokine receptor type 2 (CCR2) expressed in the brain are involved in various neuroinflammatory disorders, such as multiple sclerosis (MS), Alzheimer's disease (AD) and Parkinson's disease (PD). However, the role of MCP-1/CCR2 in alcohol-induced brain damage is unclear. Recent evidence indicates that alcohol exposure increased the activity of MCP-1/CCR2 in both mature and developing central nervous systems (CNS). MCP-1/CCR2 signaling in the brain was involved in alcohol drinking behavior. MCP-1/CCR2 inhibition alleviated alcohol neurotoxicity by reducing microglia activation/neuroinflammation in the developing brain and spinal cord. In this review, we discussed the role of MCP-1/CCR2 signaling in alcohol-induced neuroinflammation and brain damage. We also discussed the signaling cascades that are involved in the activation of MCP-1/CCR2 in response to alcohol exposure.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Jia Luo
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY 40536, USA; Lexington VA Health Care System, Research & Development, 1101 Veterans Drive, Lexington, KY 40502, USA.
| |
Collapse
|
28
|
Smith KA, Pulsipher A, Gabrielsen DA, Alt JA. Biologics in Chronic Rhinosinusitis: An Update and Thoughts for Future Directions. Am J Rhinol Allergy 2018; 32:412-423. [PMID: 30021447 DOI: 10.1177/1945892418787132] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Background Potential biologic therapies for chronic rhinosinusitis (CRS) is a growing field of interest and research. Biologics target specific immune cells or inflammatory pathways within a disease process, increasing drug efficacy while reducing complications. The success of biologics in other inflammatory conditions such as asthma and atopic dermatitis has spurred much of the corresponding research in CRS. A rapid expansion in the volume of research concerning biologic therapies with potential crossover to treating CRS has made it difficult to stay current. Furthermore, much of the literature has been focused on allergy, asthma, and immunology subspecialties. As the role for biologic therapies in CRS continues to expand, it is increasingly important for otolaryngologists to remain up to date on their progression. Objective The objectives of this review are to provide an update on the growing field of biologics for otolaryngologists who treat CRS and discuss potential future areas of research. Methods A literature review of biologic therapies studied in CRS was performed. In addition, a detailed review of all biologic therapies targeting inflammatory markers involved in Th1-, Th2-, and Th17-mediated inflammation was performed to identify potential areas for future research. The role for biologic therapies in CRS, endotypes of CRS, current biologic therapies studies in CRS, and future areas for research were reviewed. Results Sixty-nine unique biologic therapies have been developed for Th1-, Th2-, and Th17-mediated inflammation. Five biologics are currently being investigated for use in patients with CRS with nasal polyposis. Conclusions As the field of biologics continues to expand, remaining up to date on the current literature may help clinicians identify patients who may benefit from biologic therapies. In addition, ongoing research in other inflammatory disorders with shared pathophysiology to CRS may reveal other potential therapies for CRS that have not previously been investigated.
Collapse
Affiliation(s)
- Kristine A Smith
- 1 Division of Otolaryngology - Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - Abigail Pulsipher
- 1 Division of Otolaryngology - Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, Utah.,2 GlycoMira Therapeutics, Inc., Salt Lake City, Utah
| | - David A Gabrielsen
- 1 Division of Otolaryngology - Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, Utah
| | - Jeremiah A Alt
- 1 Division of Otolaryngology - Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
29
|
Absence of IL-17A in Litomosoides sigmodontis-infected mice influences worm development and drives elevated filarial-specific IFN-γ. Parasitol Res 2018; 117:2665-2675. [PMID: 29931394 PMCID: PMC6061040 DOI: 10.1007/s00436-018-5959-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 06/05/2018] [Indexed: 01/12/2023]
Abstract
Lymphatic filariasis, onchocerciasis and loiasis are widespread neglected tropical diseases causing serious public health problems and impacting the socio-economic climate in endemic communities. More than 100 million people currently suffer from filarial infections but disease-related symptoms and infection-induced immune mechanisms are still ambiguous. Although most infected individuals have dominant Th2 and regulatory immune responses leading to a homeostatic regulated state, filarial-induced overt pathology like lymphedema, dermal pathologies or blindness can occur. Interestingly, besides dominant Th2 and regulatory T cell activation, increased Th17-induced immune responses were associated with filarial infection and overt helminth-induced pathology in humans. However, the immunological mechanisms of Th17 cells and the release of IL-17A during filarial infections remain unclear. To decipher the role of IL-17A during filarial infection, we naturally infected IL-17A-/- and wildtype C57BL/6 mice with the rodent filariae Litomosoides sigmodontis and analysed parasite development and immune alterations. Our study reveals that infected IL-17A-deficient C57BL/6 mice present reduced worm burden on days 7 and 28 p.i. but had longer adult worms on day 28 p.i. in the thoracic cavity (TC), the site of infection. In addition, infiltration of CD4+ T cells, CD4+Foxp3+ regulatory T and functional CD4+Rorγt+pStat3+ Th17 cells in the TC was reduced in IL-17A-deficient mice accompanied by reduced eotaxin-1 and CCL17 levels. Furthermore, mediastinal lymph node cells isolated from IL-17A-/- mice showed increased filarial-specific IFN-γ but not IL-4, IL-6, or IL-21 secretion. This study shows that Th17 signalling is important for host immune responses against filarial infection but appears to facilitate worm growth in those that reach the TC.
Collapse
|
30
|
Chang HC, Huang PH, Syu FS, Hsieh CH, Chang SLY, Lu J, Chen HC. Critical involvement of atypical chemokine receptor CXCR7 in allergic airway inflammation. Immunology 2018; 154:274-284. [PMID: 29250768 DOI: 10.1111/imm.12881] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 11/20/2017] [Accepted: 12/03/2017] [Indexed: 12/21/2022] Open
Abstract
Trafficking and recruitment of immune cells to the site of inflammation with spatial and temporal synchronization is crucial for the development of allergic airway inflammation. Particularly, chemokines are known to be key players in these processes. Previous studies revealed that the CXCL12/CXCR4 axis plays an important role in regulating allergic airway inflammation. However, the role of CXCR7, a recently discovered second receptor for CXCL12, in regulating airway inflammation has not been explored. Initially, CXCR7 was considered as a decoy receptor; however, numerous subsequent studies revealed that engagement of CXCR7 triggered its own signalling or modulated CXCR4-mediated signalling. In the present study, we detected the expression of CXCR7 in airway epithelial cells. Use of a lentiviral delivery system to knock down the expression of CXCR7 in the lung of sensitized mice abrogated the cardinal features of asthma, indicating that CXCR7 plays a role in regulating allergic airway inflammation. The activation of mitogen-activated protein kinase and Akt signalling in response to CXCL12 in the mouse epithelial cell line MLE-12 was reduced when CXCR7 expression was knocked down. However, either knockdown or overexpression of CXCR7 in MLE-12 did not affect CXCL12-mediated calcium influx, indicating that CXCR7 does not modulate CXCR4-mediated signalling, and that it functions as a signalling receptor rather than a decoy receptor. Finally, we found that the expression of chemokine CCL2 is regulated by CXCR7/CXCL12-mediated signalling through β-arrestin in airway epithelial cells. Hence, regulating the expression of CCL2 in airway epithelial cells may be one mechanism by which CXCR7 participates in regulating allergic airway inflammation.
Collapse
Affiliation(s)
- Hung-Chih Chang
- Graduate Institute of Life Science, National Defence Medical Centre, Taipei, Taiwan
| | - Po-Han Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Fu-Sheng Syu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chia-Hung Hsieh
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Sunny Li-Yun Chang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Biomedical Informatics, Asia University, Taichung, Taiwan
| | - Jean Lu
- Graduate Institute of Life Science, National Defence Medical Centre, Taipei, Taiwan.,The Genomics Research Centre, Academia Sinica, Taipei, Taiwan.,Genome and Systems Biology Degree Programme, College of Life Science, National Taiwan University, Taipei, Taiwan.,Department of Life Science, Tzu Chi University, Hualien, Taiwan
| | - Hui-Chen Chen
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Department of Optometry, Asia University, Taichung, Taiwan.,Department of Microbiology and Immunology, School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
31
|
Lee H, Zhang D, Wu J, Otterbein LE, Jin Y. Lung Epithelial Cell-Derived Microvesicles Regulate Macrophage Migration via MicroRNA-17/221-Induced Integrin β 1 Recycling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:1453-1464. [PMID: 28674181 PMCID: PMC5561736 DOI: 10.4049/jimmunol.1700165] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/11/2017] [Indexed: 12/26/2022]
Abstract
Robust lung inflammation is one of the prominent features in the pathogenesis of acute lung injury (ALI). Macrophage migration and recruitment are often seen at the early stage of lung inflammatory responses to noxious stimuli. Using an acid inhalation-induced lung injury model, we explored the mechanisms by which acid exposure initiates macrophage recruitment and migration during development of ALI. The lung epithelium comprises a large surface area and functions as a first-line defense against noxious insults. We found that acid exposure induced a remarkable microvesicle (MV) release from lung epithelium as detected in bronchoalveolar lavage fluid. Significantly elevated RNA, rather than protein, was found in these epithelium-derived MVs after acid and included several highly elevated microRNAs, including microRNA (miR)-17 and miR-221. Acid-induced epithelial MV release promoted macrophage migration in vitro and recruitment into the lung in vivo and required, in part, MV shuttling of miR-17 and/or miR-221. Mechanistically, acid-induced epithelial MV miR-17/221 promoted β1 integrin recycling and presentation back onto the surface of macrophages, in part via a Rab11-mediated pathway. Integrin β1 is known to play an essential role in regulating macrophage migration. Taken together, acid-induced ALI results in epithelial MV shuttling of miR-17/221 that in turn modulates macrophage β1 integrin recycling, promoting macrophage recruitment and ultimately contributing to lung inflammation.
Collapse
Affiliation(s)
- Heedoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| | - Duo Zhang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| | - Jingxuan Wu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| | - Leo E Otterbein
- Department of Surgery, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA 02215
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118; and
| |
Collapse
|
32
|
Castan L, Magnan A, Bouchaud G. Chemokine receptors in allergic diseases. Allergy 2017; 72:682-690. [PMID: 27864967 DOI: 10.1111/all.13089] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2016] [Indexed: 12/21/2022]
Abstract
Under homeostatic conditions, as well as in various diseases, leukocyte migration is a crucial issue for the immune system that is mainly organized through the activation of bone marrow-derived cells in various tissues. Immune cell trafficking is orchestrated by a family of small proteins called chemokines. Leukocytes express cell-surface receptors that bind to chemokines and trigger transendothelial migration. Most allergic diseases, such as asthma, rhinitis, food allergies, and atopic dermatitis, are generally classified by the tissue rather than the type of inflammation, making the chemokine/chemokine receptor system a key point of the immune response. Moreover, because small antagonists can easily block such receptors, various molecules have been developed to suppress the recruitment of immune cells during allergic reactions, representing potential new drugs for allergies. We review the chemokines and chemokine receptors that are important in asthma, food allergies, and atopic dermatitis and their respectively developed antagonists.
Collapse
Affiliation(s)
- L. Castan
- INRA; UR1268 BIA; Nantes France
- INSERM; UMR1087; lnstitut du thorax; Nantes France
- CNRS; UMR6291; Nantes France
- Université de Nantes; Nantes France
| | - A. Magnan
- INSERM; UMR1087; lnstitut du thorax; Nantes France
- CNRS; UMR6291; Nantes France
- CHU de Nantes; Service de Pneumologie; Institut du thorax; Nantes France
| | | |
Collapse
|
33
|
Lambrecht BN, Persson EK, Hammad H. Myeloid Cells in Asthma. Microbiol Spectr 2017; 5:10.1128/microbiolspec.mchd-0053-2016. [PMID: 28102118 PMCID: PMC11687443 DOI: 10.1128/microbiolspec.mchd-0053-2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Indexed: 12/24/2022] Open
Abstract
Asthma is a heterogeneous chronic inflammatory disorder of the airways, and not surprisingly, many myeloid cells play a crucial role in pathogenesis. Antigen-presenting dendritic cells are the first to recognize the allergens, pollutants, and viruses that are implicated in asthma pathogenesis, and subsequently initiate the adaptive immune response by migrating to lymph nodes. Eosinophils are the hallmark of type 2 inflammation, releasing toxic compounds in the airways and contributing to airway remodeling. Mast cells and basophils control both the early- and late-phase allergic response and contribute to alterations in smooth muscle reactivity. Finally, relatively little is known about neutrophils and macrophages in this disease. Although many of these myeloid cells respond well to treatment with inhaled steroids, there is now an increasing armamentarium of targeted biologicals that can specifically eliminate only one myeloid cell population, like eosinophils. It is only with those new tools that we will be able to fully understand the role of myeloid cells in chronic asthma in humans.
Collapse
Affiliation(s)
- Bart N Lambrecht
- VIB Center for Inflammation Research, Ghent University, 9000 Gent, Belgium
- Department of Pulmonary Medicine, Ghent University Hospital, 9000 Gent, Belgium
| | - Emma K Persson
- VIB Center for Inflammation Research, Ghent University, 9000 Gent, Belgium
| | - Hamida Hammad
- VIB Center for Inflammation Research, Ghent University, 9000 Gent, Belgium
- Department of Pulmonary Medicine, Ghent University Hospital, 9000 Gent, Belgium
| |
Collapse
|
34
|
Liang Z, Nie H, Xu Y, Peng J, Zeng Y, Wei Y, Wen X, Qiu J, Zhong W, Deng X, He J. Therapeutic effects of rosmarinic acid on airway responses in a murine model of asthma. Int Immunopharmacol 2016; 41:90-97. [PMID: 27825045 DOI: 10.1016/j.intimp.2016.10.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 10/06/2016] [Accepted: 10/13/2016] [Indexed: 12/22/2022]
Abstract
Rosmarinic acid (RA) is an active component of a traditional Chinese herbal medicine. Previously, we reported that RA exerted a strong anti-inflammatory effect in a mouse acute lung injury model. Therefore, we hypothesized that RA might also have potential therapeutic effects in a murine model of asthma. In this study, we aimed to evaluate the anti-asthmatic activity of RA and explored its possible molecular mechanisms of action. Female BALB/c mice that had been sensitized to and challenged with ovalbumin (Ova) were treated with RA (20mg/kg) 1h after challenge. The results showed that RA greatly diminished the number of inflammatory cells and the production of Th2 cytokines in the bronchoalveolar lavage fluid (BALF); significantly reduced the secretion of total IgE, Ova-specific IgE, and eotaxin; and markedly ameliorated airway hyperresponsiveness (AHR) compared with Ova-induced mice. Histological studies further revealed that RA substantially decreased inflammatory cells infiltration and mucus hypersecretion compared with Ova-induced mice. Moreover, our results suggested that the protective effects of RA were mediated by the inhibition of JNK and p38 MAPK phosphorylation and nuclear factor-κB (NF-κB) activation. Furthermore, RA treatment resulted in a significant reduction in the mRNA expression of AMCase, CCL11, CCR3, Ym2 and E-selectin in lung tissue. These findings suggest that RA may effectively delay the development of airway inflammation and could thus be used as a therapy for allergic asthma.
Collapse
Affiliation(s)
- Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530005, PR China
| | - Haiying Nie
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530005, PR China
| | - Yangfeng Xu
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530005, PR China
| | - Jianbo Peng
- Guangxi Engineering Research Center for Veterinary Medicine, Nanning, Guangxi 530007, PR China
| | - Yun Zeng
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530005, PR China
| | - Yingyi Wei
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530005, PR China
| | - Xuemei Wen
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530005, PR China
| | - Jiaming Qiu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062, PR China
| | - Weiting Zhong
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062, PR China
| | - Xuming Deng
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin 130062, PR China
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi 530005, PR China.
| |
Collapse
|
35
|
Liang Z, Xu Y, Wen X, Nie H, Hu T, Yang X, Chu X, Yang J, Deng X, He J. Rosmarinic Acid Attenuates Airway Inflammation and Hyperresponsiveness in a Murine Model of Asthma. Molecules 2016; 21:E769. [PMID: 27304950 PMCID: PMC6274450 DOI: 10.3390/molecules21060769] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/02/2016] [Accepted: 06/08/2016] [Indexed: 01/05/2023] Open
Abstract
Rosmarinic acid (RA) has numerous pharmacologic effects, including anti-oxidant, anti-inflammatory, and analgesic effects. This study aimed to evaluate the preventive activity of RA in a murine model of asthma and to investigate its possible molecular mechanisms. Female BALB/c mice sensitized and challenged with ovalbumin (Ova) were pretreated with RA (5, 10 or 20 mg/kg) at 1 h before Ova challenge. The results demonstrated that RA markedly inhibited increases in inflammatory cells and Th2 cytokines in the bronchoalveolar lavage fluid (BALF), significantly reduced the total IgE and Ova-specific IgE concentrations, and greatly ameliorated airway hyperresponsiveness (AHR) compared with the control Ova-induced mice. Histological analyses showed that RA substantially decreased the number of inflammatory cells and mucus hypersecretion in the airway. In addition, our results suggested that the protective effects of RA might be mediated by the suppression of ERK, JNK and p38 phosphorylation and activation of nuclear factor-κB (NF-κB). Furthermore, RA pretreatment resulted in a noticeable reduction in AMCase, CCL11, CCR3, Ym2 and E-selectin mRNA expression in lung tissues. These findings suggest that RA may effectively delay the progression of airway inflammation.
Collapse
Affiliation(s)
- Zhengmin Liang
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China.
| | - Yangfeng Xu
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China.
| | - Xuemei Wen
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China.
| | - Haiying Nie
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China.
| | - Tingjun Hu
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China.
| | - Xiaofeng Yang
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, Jilin, China.
| | - Xiao Chu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, Jilin, China.
| | - Jian Yang
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China.
| | - Xuming Deng
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, Jilin, China.
| | - Jiakang He
- College of Animal Science and Technology, Guangxi University, Nanning 530005, China.
| |
Collapse
|
36
|
Abstract
Leukocyte infiltration of the lung is a characteristic feature of allergic asthma and it is thought that these cells are selectively recruited by chemokines. Extensive research has confirmed that chemokine receptors are expressed on the main cell types involved in asthma, including eosinophils, T helper type 2 cells, mast cells and even neutrophils. Moreover, animal experiments have outlined a functional role for these receptors and their ligands. Chemokines signal via seven-transmembrane spanning G-protein coupled receptors, which are favored targets of the pharmaceutical industry due to the possibility of designing small-molecule inhibitors. In fact, this family represents the first group of cytokines where small-molecule inhibitors have been designed. However, the search for efficient antagonists of chemokine/chemokine receptors has not been easy; a particular feature of the chemokine system is the number of molecules with overlapping functions and binding specificities, as well as the difficulty in reconciling the in vivo biologic functional validation of chemokines in rodent models with the development of antagonists which bind the human receptor, because of the lack of species cross-reactivity. The chemokines and their receptors that are active during allergic reactions are reviewed. Possible points of interaction that may be a target for development of new therapies, as well as the progress to date in developing inhibitors of key chemokine receptors for asthma therapy, are also discussed.
Collapse
Affiliation(s)
- Clare M Lloyd
- Leukocyte Biology Section, NHLI, Faculty of Medicine, Imperial College, London, England
| | | |
Collapse
|
37
|
Xu Y, Chu N, Qiu X, Gober HJ, Li D, Wang L. The interconnected role of chemokines and estrogen in bone metabolism. Biosci Trends 2016; 10:433-444. [DOI: 10.5582/bst.2016.01072] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Yingping Xu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| | - Nan Chu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
| | - Xuemin Qiu
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| | | | - Dajin Li
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| | - Ling Wang
- Obstetrics and Gynecology Hospital of Fudan University
- Shanghai Key Laboratory of Female Reproductive Endocrine-related Diseases
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, IBS, Shanghai Medical College, Fudan University
- The Academy of Integrative Medicine of Fudan University
| |
Collapse
|
38
|
Park K, Lee JS, Choi JS, Nam YJ, Han JH, Byun HD, Song MJ, Oh JS, Kim SG, Choi Y. Identification and Characterization of Baicalin as a Phosphodiesterase 4 Inhibitor. Phytother Res 2015; 30:144-51. [PMID: 26549702 DOI: 10.1002/ptr.5515] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 10/06/2015] [Accepted: 10/16/2015] [Indexed: 01/04/2023]
Abstract
Asthma is a chronic inflammatory disease of lung airways, and pharmacological inhibitors of cyclic adenosine monophosphate-specific phosphodiesterase 4 (PDE4) have been considered as therapeutics for the treatment of asthma. However, development of PDE4 inhibitors in clinical trials has been hampered because of the severe side effects of non-selective PDE4 inhibitors. Here, screening of a plant extract library in conjunction with dereplication technology led to identification of baicalin as a new type of PDE4-selective inhibitor. We demonstrated that while rolipram inhibited the enzyme activity of a range of PDE4 subtypes in in vitro enzyme assays, baicalin selectively inhibited the enzyme activity of PDE4A and 4B. In addition, baicalin suppressed lipopolysaccharide-induced TNF-α expression in macrophage where PDE4B plays a key role in lipopolysaccharide-induced signaling. Furthermore, baicalin treatment in an animal model of allergic asthma reduced inflammatory cell infiltration and TNF-α levels in bronchoalveolar lavage fluids, indicating that the antiinflammatory effects of baicalin in vivo are attributable, in part, to its ability to inhibit PDE4.
Collapse
Affiliation(s)
- Kyuhee Park
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi-do, 443-270, Korea
| | - Jong Suk Lee
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi-do, 443-270, Korea
| | - Jung Suk Choi
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi-do, 443-270, Korea
| | - Yeon-Ju Nam
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi-do, 443-270, Korea
| | - Jong-Heon Han
- Bio Convergence Center, Jeju Technopark, Jeju city, Jeju-do, 699-121, Korea
| | - Hoo-Dhon Byun
- Bio Convergence Center, Jeju Technopark, Jeju city, Jeju-do, 699-121, Korea
| | - Myung-Jin Song
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi-do, 443-270, Korea
| | - Joa-Sup Oh
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi-do, 443-270, Korea
| | - Sung Gyu Kim
- Bio Convergence Center, Jeju Technopark, Jeju city, Jeju-do, 699-121, Korea
| | - Yongmun Choi
- Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi-do, 443-270, Korea
| |
Collapse
|
39
|
Shukla A, Mishra A, Venkateshaiah SU, Manohar M, Mahadevappa CP, Mishra A. Elements Involved In Promoting Eosinophilic Gastrointestinal Disorders. ACTA ACUST UNITED AC 2015; 6. [PMID: 27840774 PMCID: PMC5102338 DOI: 10.4172/2157-7412.1000265] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Eosinophilic gastrointestinal disorders (EGID) are food allergen-induced allergic gastrointestinal disorders, characterized by accumulation of highly induced eosinophils in different segments of gastrointestinal tract along with eosinophil microabssess and extracellular eosinophilic granules in the epithelial layer. EGID are both IgE- and cell-mediated group of diseases that include eosinophilic esophagitis (EoE), eosinophilic gastritis (EG), eosinophilic gastroenteritis (EGE) and eosinophilic colitis (EC). Despite the increased incidences and considerable progress made in understanding EGID pathogenesis. The mechanism is still not well understood. It has been shown that IL-4, IL-5, IL-13, IL-15, IL-18, eotaxin-1, eotaxin-2 and eotaxin-3 play a critical role in EGID pathogenesis. Currently, the only criterion for diagnosing EoE, EGE and EC are repetitive endoscopic and histopathological evaluation of biopsies along with other clinical characteristics/manifestations. Antigen elimination and corticosteroid therapies are the most effective therapies currently in practice for the treatment of EGID. The cytokines (anti-IL-5 and anti-IL-13) therapy trials were not very successful in case of EoE. Most recently, a clinical trial using anti-IL-13 reported only 60% reduced esophageal eosinophilia without achieving primary endpoint. This clinical finding is not surprising and is in accordance with our earlier report indicating that IL-13 is not critical in the initiation of EoE. Notably, EGID still has no reliable noninvasive diagnostic biomarkers. Hence, there is a great necessity to identify novel noninvasive diagnostic biomarkers that can easily diagnose EGID and provide an effective therapy. Now, the attention is required to target cell types like iNKT cells that produce eosinophil active cytokines and is found induced in the pathogenesis of both experimental and human EoE. iNKT cell neutralization is shown to protect allergen-induced EoE in experimental model. In this review, we have discussed the key elements that are critical in the disease initiation, progression, pathogenesis and important for future diagnostic and therapeutic interventions for EGID.
Collapse
Affiliation(s)
- Anshi Shukla
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, 1430 Tulane Avenue, New Orleans, LA 70112
| | - Akanksha Mishra
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, 1430 Tulane Avenue, New Orleans, LA 70112
| | | | - Murli Manohar
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, 1430 Tulane Avenue, New Orleans, LA 70112
| | | | - Anil Mishra
- Department of Medicine, Section of Pulmonary Diseases, Tulane Eosinophilic Disorder Center, 1430 Tulane Avenue, New Orleans, LA 70112
| |
Collapse
|
40
|
Nau F, Miller J, Saravia J, Ahlert T, Yu B, Happel KI, Cormier SA, Nichols CD. Serotonin 5-HT₂ receptor activation prevents allergic asthma in a mouse model. Am J Physiol Lung Cell Mol Physiol 2015; 308:L191-8. [PMID: 25416380 DOI: 10.1152/ajplung.00138.2013] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Asthma is an inflammatory disease of the lung characterized by airways hyper-responsiveness (AHR), inflammation, and mucus hyperproduction. Current mainstream therapies include bronchodilators that relieve bronchoconstriction and inhaled glucocorticoids to reduce inflammation. The small molecule hormone and neurotransmitter serotonin has long been known to be involved in inflammatory processes; however, its precise role in asthma is unknown. We have previously established that activation of serotonin 5-hydroxytryptamine (5-HT)(2A) receptors has potent anti-inflammatory activity in primary cultures of vascular tissues and in the whole animal in vasculature and gut tissues. The 5-HT(2A) receptor agonist, (R)-2,5-dimethoxy-4-iodoamphetamine [(R)-DOI] is especially potent. In this work, we have examined the effect of (R)-DOI in an established mouse model of allergic asthma. In the ovalbumin mouse model of allergic inflammation, we demonstrate that inhalation of (R)-DOI prevents the development of many key features of allergic asthma, including AHR, mucus hyperproduction, airways inflammation, and pulmonary eosinophil recruitment. Our results highlight a likely role of the 5-HT2 receptors in allergic airways disease and suggest that 5-HT2 receptor agonists may represent an effective and novel small molecule-based therapy for asthma.
Collapse
|
41
|
Xiong Y, Wang J, Yu H, Zhang X, Miao C. Anti-asthma potential of crocin and its effect on MAPK signaling pathway in a murine model of allergic airway disease. Immunopharmacol Immunotoxicol 2015; 37:236-43. [PMID: 25753844 DOI: 10.3109/08923973.2015.1021356] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Crocin, a diterpenoid glucoside, has multitudinous activities such as anti-inflammation, anti-allergy, anti-oxidation and relaxing smooth muscles. OBJECTIVE In this study, the potential of crocin as an anti-asthma agent was investigated in a murine model. MATERIALS AND METHODS BALB/c mice were sensitized and challenged by ovalbumin (OVA) to induce allergic airway inflammation, with crocin administered one hour before every OVA challenge. Airway hyper-reactivity was evaluated by lung function analysis systems. Leukocyte counts in bronchoalveolar lavage fluid (BALF) were measured by a hemocytometer and Diff-Quick-stained smears. Lung tissues were stained with hematoxylin-eosin, Congo red and methylene blue for histopathological inspection. Inflammatory mediators in serum, BALF and lung were measured by ELISA or RT-PCR. Effects of crocin on MAPK signaling pathways were investigated by western blot analysis. RESULTS Crocin significantly suppressed airway inflammation and hyper-reactivity, reduced levels of BALF interleukin (IL-4), IL-5, IL-13 and tryptase, lung eosinophil peroxidase and serum OVA-specific IgE, and inhibited the expression of lung eotaxin, p-ERK, p-JNK and p-p38 in the OVA-challenged mice. CONCLUSIONS These results demonstrated that the suppression of crocin on airway inflammation and hyper-reactivity in a murine model, thus crocin might have a great potential to be a candidate for the treatment of asthma.
Collapse
Affiliation(s)
- Youyi Xiong
- College of Food and Drug, Anhui Science and Technology University , Fengyang, Anhui , China and
| | | | | | | | | |
Collapse
|
42
|
Wang A, Wang Z, Cao Y, Cheng S, Chen H, Bunjhoo H, Xie J, Wang C, Xu Y, Xiong W. CCL2/CCR2-dependent recruitment of Th17 cells but not Tc17 cells to the lung in a murine asthma model. Int Arch Allergy Immunol 2015; 166:52-62. [PMID: 25765592 DOI: 10.1159/000371764] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 12/19/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Interleukin (IL)-17 has been implicated in the pathogenesis of asthma and the progression of airway inflammation. Here, we used a model of allergic asthma and found that the frequencies of IL-17-secreting T helper (Th)17 and CD8 (Tc)17 cells were both significantly increased, as was the expression of the CC chemokine receptor (CCR2) on the surface of these cells. CC chemokine ligand 2 (CCL2) has been shown to mediate the activation and recruitment of inflammatory cells in asthma, which are also skewed after ovalbumin (OVA) challenge. However, the role of CCL2 on Th17 cells and Tc17 cells in asthma has not been illuminated. METHODS Mice that were sensitized and challenged with OVA received anti-CCL2 antibody (Ab; 5 μg/day intratracheally) or CCR2 antagonist (RS504393, 2 mg/kg/day intraperitoneally) prior to the challenge. Some mice received an isotype control Ab or vehicle alone. We then assessed the effects of allergic asthma and anti-CCL2 Ab or CCR2 antagonist treatment on the levels of IL-17 and CCL2, the Th17 and Tc17 cell frequencies and lung tissue inflammation. RESULTS We demonstrated that CCL2 and IL-17 levels and the frequency of Th17 and Tc17 cells in lung tissues and bronchoalveolar lavage fluid increased in the asthma group compared with the normal control mice. Blocking the CCL2/CCR2 axis greatly reduced the Th17 but not the Tc17 cell frequency, and revealed a suppressive effect on airway inflammation. CONCLUSION These findings indicate a role for the CCL2/CCR2 axis in mediating Th17 but not Tc17 cell migration during acute allergic airway inflammation.
Collapse
Affiliation(s)
- Aili Wang
- Department of Respiratory and Critical Care Medicine, Tongji Hospital and Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Martinu T, Gowdy KM, Nugent JL, Sun J, Kinnier CV, Nelson ME, Lyes MA, Kelly FL, Foster WM, Gunn MD, Palmer SM. Role of C-C motif ligand 2 and C-C motif receptor 2 in murine pulmonary graft-versus-host disease after lipopolysaccharide inhalations. Am J Respir Cell Mol Biol 2015; 51:810-21. [PMID: 24921973 DOI: 10.1165/rcmb.2013-0451oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Environmental exposures are a potential trigger of chronic pulmonary graft-versus-host disease (pGVHD) after successful recovery from hematopoietic cell transplant (HCT). We hypothesized that inhalations of LPS, a prototypic environmental stimulus, trigger pGVHD via increased pulmonary recruitment of donor-derived antigen-presenting cells (APCs) through the C-C motif ligand 2 (CCL2)-C-C motif receptor 2 (CCR2) chemokine axis. B10.BR(H2(k)) and C57BL/6(H2(b)) mice underwent allogeneic (Allo) or syngeneic (Syn) HCT with wild-type (WT) C57BL/6, CCL2(-/-), or CCR2(-/-) donors. After 4 weeks, recipient mice received daily inhaled LPS for 5 days and were killed at multiple time points. Allo mice exposed to repeated inhaled LPS developed prominent lymphocytic bronchiolitis, similar to human pGVHD. The increase in pulmonary T cells in Allo mice after LPS exposures was accompanied by increased CCL2, CCR2, and Type-1 T-helper cytokines as well as by monocytes and monocyte-derived dendritic cells (moDCs) compared with Syn and nontransplanted controls. Using CCL2(-/-) donors leads to a significant decrease in lung DCs but to only mildly reduced CD4 T cells. Using CCR2(-/-) donors significantly reduces lung DCs and moDCs but does not change T cells. CCL2 or CCR2 deficiency does not alter pGVHD pathology but increases airway hyperreactivity and IL-5 or IL-13 cytokines. Our results show that hematopoietic donor-derived CCL2 and CCR2 regulate recruitment of APCs to the Allo lung after LPS exposure. Although they do not alter pathologic pGVHD, their absence is associated with increased airway hyperreactivity and IL-5 and IL-13 cytokines. These results suggest that the APC changes that result from CCL2-CCR2 blockade may have unexpected effects on T cell differentiation and physiologic outcomes in HCT.
Collapse
|
44
|
Mucosal Eosinophils. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00044-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Phospholipase cε, an effector of ras and rap small GTPases, is required for airway inflammatory response in a mouse model of bronchial asthma. PLoS One 2014; 9:e108373. [PMID: 25269075 PMCID: PMC4182471 DOI: 10.1371/journal.pone.0108373] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 08/20/2014] [Indexed: 11/29/2022] Open
Abstract
Background Phospholipase Cε (PLCε) is an effector of Ras and Rap small GTPases and expressed in non-immune cells. It is well established that PLCε plays an important role in skin inflammation, such as that elicited by phorbol ester painting or ultraviolet irradiation and contact dermatitis that is mediated by T helper (Th) 1 cells, through upregulating inflammatory cytokine production by keratinocytes and dermal fibroblasts. However, little is known about whether PLCε is involved in regulation of inflammation in the respiratory system, such as Th2-cells-mediated allergic asthma. Methods We prepared a mouse model of allergic asthma using PLCε+/+ mice and PLCεΔX/ΔX mutant mice in which PLCε was catalytically-inactive. Mice with different PLCε genotypes were immunized with ovalbumin (OVA) followed by the challenge with an OVA-containing aerosol to induce asthmatic response, which was assessed by analyzing airway hyper-responsiveness, bronchoalveolar lavage fluids, inflammatory cytokine levels, and OVA-specific immunoglobulin (Ig) levels. Effects of PLCε genotype on cytokine production were also examined with primary-cultured bronchial epithelial cells. Results After OVA challenge, the OVA-immunized PLCεΔX/ΔX mice exhibited substantially attenuated airway hyper-responsiveness and broncial inflammation, which were accompanied by reduced Th2 cytokine content in the bronchoalveolar lavage fluids. In contrast, the serum levels of OVA-specific IgGs and IgE were not affected by the PLCε genotype, suggesting that sensitization was PLCε-independent. In the challenged mice, PLCε deficiency reduced proinflammatory cytokine production in the bronchial epithelial cells. Primary-cultured bronchial epithelial cells prepared from PLCεΔX/ΔX mice showed attenuated pro-inflammatory cytokine production when stimulated with tumor necrosis factor-α, suggesting that reduced cytokine production in PLCεΔX/ΔX mice was due to cell-autonomous effect of PLCε deficiency. Conclusions PLCε plays an important role in the pathogenesis of bronchial asthma through upregulating inflammatory cytokine production by the bronchial epithelial cells.
Collapse
|
46
|
Singh SR, Sutcliffe A, Kaur D, Gupta S, Desai D, Saunders R, Brightling CE. CCL2 release by airway smooth muscle is increased in asthma and promotes fibrocyte migration. Allergy 2014; 69:1189-97. [PMID: 24931417 PMCID: PMC4215601 DOI: 10.1111/all.12444] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2014] [Indexed: 11/28/2022]
Abstract
BACKGROUND Asthma is characterized by variable airflow obstruction, airway inflammation, airway hyper-responsiveness and airway remodelling. Airway smooth muscle (ASM) hyperplasia is a feature of airway remodelling and contributes to bronchial wall thickening. We sought to investigate the expression levels of chemokines in primary cultures of ASM cells from asthmatics vs healthy controls and to assess whether differentially expressed chemokines (i) promote fibrocyte (FC) migration towards ASM and (ii) are increased in blood from subjects with asthma and in sputum samples from those asthmatics with bronchial wall thickening. METHODS Chemokine concentrations released by primary ASM were measured by MesoScale Discovery platform. The chemokine most highly expressed by ASM from asthmatics compared with healthy controls was confirmed by ELISA, and expression of its cognate chemokine receptor by FCs was examined by immunofluorescence and flow cytometry. The role of this chemokine in FC migration towards ASM was investigated by chemotaxis assays. RESULTS Chemokine (C-C motif) ligand 2 (CCL2) levels were increased in primary ASM supernatants from asthmatics compared with healthy controls. CCR2 was expressed on FCs. Fibrocytes migrated towards recombinant CCL2 and ASM supernatants. These effects were inhibited by CCL2 neutralization. CCL2 levels were increased in blood from asthmatics compared with healthy controls, and sputum CCL2 was increased in asthmatics with bronchial wall thickening. CONCLUSIONS Airway smooth muscle-derived CCL2 mediates FC migration and potentially contributes to the development of ASM hyperplasia in asthma.
Collapse
Affiliation(s)
- S R Singh
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of LeicesterLeicester, UK
| | - A Sutcliffe
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of LeicesterLeicester, UK
| | - D Kaur
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of LeicesterLeicester, UK
| | - S Gupta
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of LeicesterLeicester, UK
| | - D Desai
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of LeicesterLeicester, UK
| | - R Saunders
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of LeicesterLeicester, UK
| | - C E Brightling
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of LeicesterLeicester, UK
| |
Collapse
|
47
|
Glista-Baker EE, Taylor AJ, Sayers BC, Thompson EA, Bonner JC. Nickel nanoparticles cause exaggerated lung and airway remodeling in mice lacking the T-box transcription factor, TBX21 (T-bet). Part Fibre Toxicol 2014; 11:7. [PMID: 24499286 PMCID: PMC3931667 DOI: 10.1186/1743-8977-11-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 02/02/2014] [Indexed: 01/22/2023] Open
Abstract
Background Nickel nanoparticles (NiNPs) are increasingly used in a variety of industrial applications, including the manufacturing of multi-walled carbon nanotubes (MWCNTs). While occupational nickel exposure is a known cause of pulmonary alveolitis, fibrosis, and cancer, the health risks of NiNPs are not well understood, especially in susceptible individuals such as asthmatics. The T-box transcription factor Tbx21 (T-bet) maintains Th1 cell development and loss of T-bet is associated with a shift towards Th2 type allergic airway inflammation that characterizes asthma. The purpose of this study was to determine the role of T-bet in susceptibility to lung remodeling by NiNPs or MWCNTs. Methods Wild-type (WT) and T-bet-/- mice were exposed to NiNPs or MWCNTs (4 mg/kg) by oropharyngeal aspiration (OPA). Necropsy was performed at 1 and 21 days. Bronchoalveolar lavage fluid (BALF) was collected for differential counting of inflammatory cells and for measurement of cytokines by ELISA. The left lung was collected for histopathology. The right lung was analyzed for cytokine or mucin (MUC5AC and MUC5B) mRNAs. Results Morphometry of alcian-blue/periodic acid Schiff (AB/PAS)-stained lung tissue showed that NiNPs significantly increased mucous cell metaplasia in T-bet-/- mice at 21 days (p < 0.001) compared to WT mice, and increased MUC5AC and MUC5B mRNAs (p < 0.05). MWCNTs also increased mucous cell metaplasia in T-bet-/- mice, but to a lesser extent than NiNPs. Chronic alveolitis was also increased by NiNPs, but not MWCNTs, in T-bet-/- mice compared to WT mice at 21 days (P < 0.001). NiNPs also increased IL-13 and eosinophils (p < 0.001) in BALF from T-bet-/- mice after 1 day. Interestingly, the chemokine CCL2 in the BALF of T-bet-/- mice was increased at 1 and 21 days (p < 0.001 and p < 0.05, respectively) by NiNPs, and to a lesser extent by MWCNTs at 1 day. Treatment of T-bet-/- mice with a monoclonal anti-CCL2 antibody enhanced NiNP-induced mucous cell metaplasia and MUC5AC mRNA levels (p < 0.05), yet marginally reduced NiNP-induced alveolitis. Conclusion These findings identify T-bet as a potentially important susceptibility factor for NiNP exposure and to a lesser extent for MWCNT exposure, and suggests that individuals with asthma are at greater risk.
Collapse
Affiliation(s)
| | | | | | | | - James C Bonner
- Environmental & Molecular Toxicology Program, Department of Biological Sciences, North Carolina State University, Campus Box 7633, Raleigh, North Carolina 27695, USA.
| |
Collapse
|
48
|
Abstract
INTRODUCTION Chemokines play important roles in inflammation and in immune responses. This article will discuss the current literature on the C-C chemokine ligand 5 (CCL5), and whether it is a therapeutic target in the context of various allergic, autoimmune or infectious diseases. AREAS COVERED Small-molecule inhibitors, chemokine and chemokine receptor-deficient mice, antibodies and modified chemokines are the current tools available for CCL5 research, and there are several ongoing clinical trials targeting the CCL5 receptors, CCR1, CCR3 and CCR5. There are fewer studies specifically targeting the chemokine itself and clinical studies with anti-CCL5 antibodies are still to be carried out. EXPERT OPINION Although clinical trials are strongly biased toward HIV treatment and prevention with blockers of CCR5, the therapeutic potential for CCL5 and its receptors in other diseases is relevant. Overall, it is not likely that specific targeting of CCL5 will result in new adjunct strategies for the treatment of infectious diseases with a major inflammatory component. However, targeting CCL5 could result in novel therapies for chronic inflammatory diseases, where it may decrease inflammatory responses and fibrosis, and certain solid tumors, where it may have a role in angiogenesis.
Collapse
Affiliation(s)
- Rafael Elias Marques
- Immunopharmacology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais,
Belo Horizonte, Brazil55 31 34092649;
| | - Rodrigo Guabiraba
- Institute of Infection, Immunity and Inflammation, University of Glasgow,
Glasgow, Scotland E-mail:
| | - Remo Castro Russo
- Immunopharmacology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais,
Belo Horizonte, Brazil55 31 34092649;
- Laboratory of Immunology and Pulmonary Mechanics, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais,
Belo Horizonte, Brazil55 31 34092938 E-mail:
| | - Mauro Martins Teixeira
- Immunopharmacology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais,
Belo Horizonte, Brazil55 31 34092649;
| |
Collapse
|
49
|
Antileukotriene reverts the early effects of inflammatory response of distal parenchyma in experimental chronic allergic inflammation. BIOMED RESEARCH INTERNATIONAL 2013; 2013:523761. [PMID: 24151607 PMCID: PMC3787560 DOI: 10.1155/2013/523761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 07/23/2013] [Accepted: 07/24/2013] [Indexed: 01/10/2023]
Abstract
Aims. Compare the effects of montelukast or dexamethasone in distal lung parenchyma and airway walls of guinea pigs (GP) with chronic allergic inflammation. Methods. GP have inhaled ovalbumin (OVA group-2x/week/4weeks). After the 4th inhalation, GP were treated with montelukast or dexamethasone. After 72 hours of the 7th inhalation, GP were anesthetised, and lungs were removed and submitted to histopathological evaluation. Results. Montelukast and dexamethasone treatments reduced the number of eosinophils in airway wall and distal lung parenchyma compared to OVA group (P < 0.05). On distal parenchyma, both treatments were effective in reducing RANTES, NF-κB, and fibronectin positive cells compared to OVA group (P < 0.001). Montelukast was more effective in reducing eotaxin positive cells on distal parenchyma compared to dexamethasone treatment (P < 0.001), while there was a more expressive reduction of IGF-I positive cells in OVA-D group (P < 0.001). On airway walls, montelukast and dexamethasone were effective in reducing IGF-I, RANTES, and fibronectin positive cells compared to OVA group (P < 0.05). Dexamethasone was more effective in reducing the number of eotaxin and NF-κB positive cells than Montelukast (P < 0.05). Conclusions. In this animal model, both treatments were effective in modulating allergic inflammation and remodeling distal lung parenchyma and airway wall, contributing to a better control of the inflammatory response.
Collapse
|
50
|
Ullah M, Eucker J, Sittinger M, Ringe J. Mesenchymal stem cells and their chondrogenic differentiated and dedifferentiated progeny express chemokine receptor CCR9 and chemotactically migrate toward CCL25 or serum. Stem Cell Res Ther 2013; 4:99. [PMID: 23958031 PMCID: PMC3854782 DOI: 10.1186/scrt310] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 06/10/2013] [Accepted: 08/12/2013] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Guided migration of chondrogenically differentiated cells has not been well studied, even though it may be critical for growth, repair, and regenerative processes. The chemokine CCL25 is believed to play a critical role in the directional migration of leukocytes and stem cells. To investigate the motility effect of serum- or CCL25-mediated chemotaxis on chondrogenically differentiated cells, mesenchymal stem cells (MSCs) were induced to chondrogenic lineage cells. METHODS MSC-derived chondrogenically differentiated cells were characterized for morphology, histology, immunohistochemistry, quantitative polymerase chain reaction (qPCR), surface profile, and serum- or CCL25-mediated cell migration. Additionally, the chemokine receptor, CCR9, was examined in different states of MSCs. RESULTS The chondrogenic differentiated state of MSCs was positive for collagen type II and Alcian blue staining, and showed significantly upregulated expression of COL2A1and SOX9, and downregulated expression of CD44, CD73, CD90, CD105 and CD166, in contrast to the undifferentiated and dedifferentiated states of MSCs. For the chondrogenic differentiated, undifferentiated, and dedifferentiated states of MSCs, the serum-mediated chemotaxis was in a percentage ratio of 33%:84%:85%, and CCL25-mediated chemotaxis was in percentage ratio of 12%:14%:13%, respectively. On the protein level, CCR9, receptor of CCL25, was expressed in the form of extracellular and intracellular domains. On the gene level, qPCR confirmed the expression of CCR9 in different states of MSCs. CONCLUSIONS CCL25 is an effective cue to guide migration in a directional way. In CCL25-mediated chemotaxis, the cell-migration rate was almost the same for different states of MSCs. In serum-mediated chemotaxis, the cell-migration rate of chondrogenically differentiated cells was significantly lower than that in undifferentiated or dedifferentiated cells. Current knowledge of the surface CD profile and cell migration could be beneficial for regenerative cellular therapies.
Collapse
Affiliation(s)
- Mujib Ullah
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jan Eucker
- Department of Hematology and Oncology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Michael Sittinger
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jochen Ringe
- Tissue Engineering Laboratory & Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|