1
|
Diez-Ahijado L, Cilleros-Portet A, Fernández-Jimenez N, Fernández MF, Guxens M, Julvez J, Llop S, Lopez-Espinosa MJ, Subiza-Pérez M, Lozano M, Ibarluzea J, Sunyer J, Bustamante M, Cosin-Tomas M. Evaluating the association between placenta DNA methylation and cognitive functions in the offspring. Transl Psychiatry 2024; 14:383. [PMID: 39304652 DOI: 10.1038/s41398-024-03094-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
The placenta plays a crucial role in protecting the fetus from environmental harm and supports the development of its brain. In fact, compromised placental function could predispose an individual to neurodevelopmental disorders. Placental epigenetic modifications, including DNA methylation, could be considered a proxy of placental function and thus plausible mediators of the association between intrauterine environmental exposures and genetics, and childhood and adult mental health. Although neurodevelopmental disorders such as autism spectrum disorder have been investigated in relation to placenta DNA methylation, no studies have addressed the association between placenta DNA methylation and child's cognitive functions. Thus, our goal here was to investigate whether the placental DNA methylation profile measured using the Illumina EPIC array is associated with three different cognitive domains (namely verbal score, perceptive performance score, and general cognitive score) assessed by the McCarthy Scales of Children's functions in childhood at age 4. To this end, we conducted epigenome-wide association analyses, including data from 255 mother-child pairs within the INMA project, and performed a follow-up functional analysis to help the interpretation of the findings. After multiple-testing correction, we found that methylation at 4 CpGs (cg1548200, cg02986379, cg00866476, and cg14113931) was significantly associated with the general cognitive score, and 2 distinct differentially methylated regions (DMRs) (including 27 CpGs) were significantly associated with each cognitive dimension. Interestingly, the genes annotated to these CpGs, such as DAB2, CEP76, PSMG2, or MECOM, are involved in placenta, fetal, and brain development. Moreover, functional enrichment analyses of suggestive CpGs (p < 1 × 10-4) revealed gene sets involved in placenta development, fetus formation, and brain growth. These findings suggest that placental DNA methylation could be a mechanism contributing to the alteration of important pathways in the placenta that have a consequence on the offspring's brain development and cognitive function.
Collapse
Affiliation(s)
- Laia Diez-Ahijado
- ISGlobal, Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Ariadna Cilleros-Portet
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU) and Biocruces-Bizkaia Health Research Institute, Basque Country, Spain
| | - Nora Fernández-Jimenez
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU) and Biocruces-Bizkaia Health Research Institute, Basque Country, Spain
| | - Mariana F Fernández
- CIBER Epidemiología y Salud Pública, Madrid, Spain
- University of Granada, Biomedical Research Centre, Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain
| | - Monica Guxens
- ISGlobal, Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
- Department of Child and Adolescent Psychiatry/Psychology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jordi Julvez
- ISGlobal, Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
- Clinical and Epidemiological Neuroscience, Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
| | - Sabrina Llop
- CIBER Epidemiología y Salud Pública, Madrid, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Public Health, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
| | - Maria-Jose Lopez-Espinosa
- CIBER Epidemiología y Salud Pública, Madrid, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Public Health, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
- Faculty of Nursing and Chiropody, University of Valencia, Valencia, Spain
| | - Mikel Subiza-Pérez
- CIBER Epidemiología y Salud Pública, Madrid, Spain
- Department of Clinical and Health Psychology and Research Methods, University of the Basque Country UPV/EHU, Avenida Tolosa 70, 20018, Donostia-San Sebastián, Spain
- Bradford Institute for Health Research, Temple Bank House, Bradford Royal Infirmary, Duckworth Lane, BD9 6RJ, Bradford, UK
- Biodonostia Health Research Institute, Group of Environmental Epidemiology and Child Development, Paseo Doctor Begiristain s/n, 20014, Donostia- San Sebastián, Spain
| | - Manuel Lozano
- Epidemiology and Environmental Health Joint Research Unit, FISABIO-Public Health, FISABIO-Universitat Jaume I-Universitat de València, Valencia, Spain
- Preventive Medicine and Public Health, Food Sciences, Toxicology and Forensic Medicine Department, Universitat de València, Valencia, Spain
| | - Jesus Ibarluzea
- CIBER Epidemiología y Salud Pública, Madrid, Spain
- Biodonostia Health Research Institute, Group of Environmental Epidemiology and Child Development, Paseo Doctor Begiristain s/n, 20014, Donostia- San Sebastián, Spain
- Ministry of Health of the Basque Government, Sub-Directorate for Public Health and Addictions of Gipuzkoa, San Sebastian, Spain
| | - Jordi Sunyer
- ISGlobal, Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Mariona Bustamante
- ISGlobal, Institute for Global Health, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Marta Cosin-Tomas
- ISGlobal, Institute for Global Health, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- CIBER Epidemiología y Salud Pública, Madrid, Spain.
| |
Collapse
|
2
|
Shimomura K, Hattori N, Iida N, Muranaka Y, Sato K, Shiraishi Y, Arai Y, Hama N, Shibata T, Narushima D, Kato M, Takamaru H, Okamoto K, Takeda H. Sleeping Beauty transposon mutagenesis identified genes and pathways involved in inflammation-associated colon tumor development. Nat Commun 2023; 14:6514. [PMID: 37845228 PMCID: PMC10579371 DOI: 10.1038/s41467-023-42228-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/29/2023] [Indexed: 10/18/2023] Open
Abstract
Chronic inflammation promotes development and progression of colorectal cancer (CRC). To comprehensively understand the molecular mechanisms underlying the development and progression of inflamed CRC, we perform in vivo screening and identify 142 genes that are frequently mutated in inflammation-associated colon tumors. These genes include senescence and TGFβ-activin signaling genes. We find that TNFα can induce stemness and activate senescence signaling by enhancing cell plasticity in colonic epithelial cells, which could act as a selective pressure to mutate senescence-related genes in inflammation-associated colonic tumors. Furthermore, we show the efficacy of the Cdk4/6 inhibitor in vivo for inflammation-associated colonic tumors. Finally, we functionally validate that Arhgap5 and Mecom are tumor suppressor genes, providing possible therapeutic targets for CRC. Thus, we demonstrate the importance of the inactivation of senescence pathways in CRC development and progression in an inflammatory microenvironment, which can help progress toward precision medicine.
Collapse
Affiliation(s)
- Kana Shimomura
- The Laboratory of Molecular Genetics, National Cancer Center Research Institute, Tokyo, Japan
| | - Naoko Hattori
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
- Department of Epigenomics, Institute for Advanced Life Sciences, Hoshi University, Tokyo, Japan
| | - Naoko Iida
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Yukari Muranaka
- The Laboratory of Molecular Genetics, National Cancer Center Research Institute, Tokyo, Japan
| | - Kotomi Sato
- The Laboratory of Molecular Genetics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuichi Shiraishi
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasuhito Arai
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Natsuko Hama
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Daichi Narushima
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo, Japan
| | - Mamoru Kato
- Division of Bioinformatics, National Cancer Center Research Institute, Tokyo, Japan
| | | | - Koji Okamoto
- Advanced Comprehensive Research Organization, Teikyo University, Tokyo, Japan
| | - Haruna Takeda
- The Laboratory of Molecular Genetics, National Cancer Center Research Institute, Tokyo, Japan.
| |
Collapse
|
3
|
Schedel M, Leach SM, Strand MJ, Danhorn T, MacBeth M, Faino AV, Lynch AM, Winn VD, Munoz LL, Forsberg SM, Schwartz DA, Gelfand EW, Hauk PJ. Molecular networks in atopic mothers impact the risk of infant atopy. Allergy 2023; 78:244-257. [PMID: 35993851 DOI: 10.1111/all.15490] [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: 03/07/2022] [Revised: 06/28/2022] [Accepted: 07/26/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND The prevalence of atopic diseases has increased with atopic dermatitis (AD) as the earliest manifestation. We assessed if molecular risk factors in atopic mothers influence their infants' susceptibility to an atopic disease. METHODS Pregnant women and their infants with (n = 174, high-risk) or without (n = 126, low-risk) parental atopy were enrolled in a prospective birth cohort. Global differentially methylated regions (DMRs) were determined in atopic (n = 92) and non-atopic (n = 82) mothers. Principal component analysis was used to predict atopy risk in children dependent on maternal atopy. Genome-wide transcriptomic analyses were performed in paired atopic (n = 20) and non-atopic (n = 15) mothers and cord blood. Integrative genomic analyses were conducted to define methylation-gene expression relationships. RESULTS Atopic dermatitis was more prevalent in high-risk compared to low-risk children by age 2. Differential methylation analyses identified 165 DMRs distinguishing atopic from non-atopic mothers. Inclusion of DMRs in addition to maternal atopy significantly increased the odds ratio to develop AD in children from 2.56 to 4.26. In atopic compared to non-atopic mothers, 139 differentially expressed genes (DEGs) were identified significantly enriched of genes within the interferon signaling pathway. Expression quantitative trait methylation analyses dependent on maternal atopy identified 29 DEGs controlled by 136 trans-acting methylation marks, some located near transcription factors. Differential expression for the same nine genes, including MX1 and IFI6 within the interferon pathway, was identified in atopic and non-atopic mothers and high-risk and low-risk children. CONCLUSION These data suggest that in utero epigenetic and transcriptomic mechanisms predominantly involving the interferon pathway may impact and predict the development of infant atopy.
Collapse
Affiliation(s)
- Michaela Schedel
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Pulmonary Medicine, University Medicine Essen-Ruhrlandklinik, Essen, Germany.,Department of Pulmonary Medicine, University Medicine Essen, University Hospital, Essen, Germany
| | - Sonia M Leach
- Department of Biomedical Research, National Jewish Health, Denver, Colorado, USA.,Center for Genes, Environment & Health, National Jewish Health, Denver, Colorado, USA
| | - Matthew J Strand
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, USA
| | - Thomas Danhorn
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, USA.,Department of Pharmacology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Morgan MacBeth
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Medical Oncology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Anna V Faino
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, Colorado, USA.,Biostatistics, Epidemiology and Research Core, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Anne M Lynch
- Department of Ophthalmology, School of Medicine, University of Colorado, Aurora, Colorado, USA.,Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Aurora, Colorado, USA.,Department of Obstetrics and Gynecology, Stanford University, Stanford, California, USA
| | - Lindsay L Munoz
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Obstetrics and Gynecology, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Shannon M Forsberg
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Department of Thoracic Oncology, University of Colorado Cancer Center, University of Colorado, Aurora, Colorado, USA
| | - David A Schwartz
- Department of Medicine, School of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Erwin W Gelfand
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Pia J Hauk
- Divisions of Allergy and Immunology and Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colorado, USA.,Section Allergy/Immunology, Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| |
Collapse
|
4
|
Gao X, Zhang W, Jia Y, Xu H, Zhu Y, Pei X. Identification of a prognosis-related ceRNA network in cholangiocarcinoma and potentially therapeutic molecules using a bioinformatic approach and molecular docking. Sci Rep 2022; 12:16247. [PMID: 36171401 PMCID: PMC9519560 DOI: 10.1038/s41598-022-20362-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 09/12/2022] [Indexed: 11/20/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a highly malignant disease with a poor prognosis, and mechanisms of initiation and development are not well characterized. It is long noncoding RNAs (lncRNAs) acting as miRNA decoys to regulate cancer-related RNAs in competing endogenous RNA (ceRNA) networks that suggest a possible molecular mechanism in CCA. The current study aims to find potential prognosis biomarkers and small molecule therapeutic targets based on the construction of a CCA prognosis-related ceRNA network. A transcriptome dataset for CCA was downloaded from the TCGA database. Differentially expressed lncRNAs (DElncRNAs), DEmiRNAs and DEmRNAs were identified based on the differential expression and a DEceRNA network was constructed using predicted miRNA-lncRNA and miRNA-mRNA interactions. Heat maps, PCA analysis, and Pathway enrichment analysis and GO enrichment analysis were conducted. The prognostic risk model and molecular docking were constructed based on identified key ceRNA networks. A DElncRNA-miRNA-mRNAs network consisting of 434 lncRNA-miRNA pairs and 284 miRNA-mRNA pairs with 200 lncRNAs, 21 miRNAs, and 245 mRNAs was constructed. There were three lncRNAs (AC090772.1, LINC00519, and THAP7-AS1) and their downstream mRNAs (MECOM, MBNL3, RCN2) screened out as prognostic factors in CAA. Three key networks (LINC00519/ hsa-mir-22/ MECOM, THAP7-AS1/hsa-mir-155/MBNL3, and THAP7-AS1/hsa-mir-155/RCN2) were identified based on binding sites prediction and survival analysis. A prognostic risk model was established with a good predictive ability (AUC = 0.66–0.83). Four anticancer small molecules, MECOM and 17-alpha-estradiol (−7.1 kcal/mol), RCN2 and emodin (−8.3 kcal/mol), RCN2 and alpha-tocopherol (−5.6 kcal/mol), and MBNL3 and 17-beta-estradiol (−7.1 kcal/mol) were identified. Based on the DEceRNA network and Kaplan–Meier survival analysis, we identified three important ceRNA networks associated with the poor prognosis of CCA. Four anti-cancer small molecules were screened out by computer-assisted drug screening as potential small molecules for the treatment of CCA. This study provides theoretical support for the development of ceRNA network-based drugs to improve the prognosis of CCA.
Collapse
Affiliation(s)
- Xiaoling Gao
- The Medical Laboratory Center, Hainan General Hospital, Haikou, 570311, China.
| | - Wenhao Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yanjuan Jia
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China.,The Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, 730000, China.,Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Hui Xu
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China.,The Institute of Clinical Research and Translational Medicine, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Yuchen Zhu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| | - Xiong Pei
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
5
|
Azar A, Bhutta MF, Del-Pozo J, Milne E, Cheeseman M. Trans-cortical vessels in the mouse temporal bulla bone are a means to recruit myeloid cells in chronic otitis media and limit peripheral leukogram changes. Front Genet 2022; 13:985214. [PMID: 36246635 PMCID: PMC9555619 DOI: 10.3389/fgene.2022.985214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic otitis media, inflammation of the middle ear, is a sequel to acute otitis media in ∼8% of children. Chronic otitis media with effusion is the most common cause of childhood deafness and is characterised by effusion of white blood cells into the auditory bulla cavity. Skull flat bones have trans-cortical vessels which are responsible for the majority of blood flow in and out of the bone. In experimental models of stroke and aseptic meningitis there is preferential recruitment of myeloid cells (neutrophils and monocytes) from the marrow in skull flat bones. We report trans-cortical vessels in the mouse temporal bone connect to the bulla mucosal vasculature and potentially represent a means to recruit myeloid cells directly into the inflamed bulla. The mutant mouse strains Junbo (MecomJbo/+) and Jeff (Fbxo11Jf/+) develop chronic otitis spontaneously; MecomJbo/+ mice have highly cellular neutrophil (90%) rich bulla exudates whereas Fbxo11Jf/+ mice have low cellularity serous effusions (5% neutrophils) indicating differing demand for neutrophil recruitment. However we found peripheral leukograms of MecomJbo/+ and Fbxo11Jf/+ mice are similar to their respective wild-type littermate controls with healthy bullae and infer preferential mobilization of myeloid cells from temporal bulla bone marrow may mitigate the need for a systemic inflammatory reaction. The cytokines, chemokines and haematopoietic factors found in the inflamed bulla represent candidate signalling molecules for myeloid cell mobilization from temporal bone marrow. The density of white blood cells in the bulla cavity is positively correlated with extent of mucosal thickening in MecomJbo/+, Fbxo11Jf/+, and EdaTa mice and is accompanied by changes in epithelial populations and bone remodelling. In MecomJbo/+ mice there was a positive correlation between bulla cavity WBC numbers and total bacterial load. The degree of inflammation varies between contralateral bullae and between mutant mice of different ages suggesting inflammation may wax and wane and may be re-initiated by a new wave of bacterial infection. Clearance of white blood cells and inflammatory stimuli from the bulla cavity is impaired and this may create a pro-inflammatory feedback loop which further exacerbates otitis media and delays its resolution.
Collapse
Affiliation(s)
- Ali Azar
- Developmental Biology Division, Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Mahmood F. Bhutta
- Brighton and Sussex Medical School, Brighton, United Kingdom
- Department of ENT, Royal Sussex County Hospital, Brighton, United Kingdom
| | - Jorge Del-Pozo
- Veterinary Pathology, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Elspeth Milne
- Veterinary Pathology, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Michael Cheeseman
- Developmental Biology Division, Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Comparative Pathology, Division of Pathology, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- *Correspondence: Michael Cheeseman,
| |
Collapse
|
6
|
Del-Pozo J, Headon DJ, Glover JD, Azar A, Schuepbach-Mallepell S, Bhutta MF, Riddell J, Maxwell S, Milne E, Schneider P, Cheeseman M. The EDA deficient mouse has Zymbal's gland hypoplasia and acute otitis externa. Dis Model Mech 2022; 15:274882. [PMID: 35107126 PMCID: PMC8990926 DOI: 10.1242/dmm.049034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 01/21/2022] [Indexed: 12/05/2022] Open
Abstract
In mice, rats, dogs and humans, the growth and function of sebaceous glands and eyelid Meibomian glands depend on the ectodysplasin signalling pathway. Mutation of genes encoding the ligand EDA, its transmembrane receptor EDAR and the intracellular signal transducer EDARADD leads to hypohidrotic ectodermal dysplasia, characterised by impaired development of teeth and hair, as well as cutaneous glands. The rodent ear canal has a large auditory sebaceous gland, the Zymbal’s gland, the function of which in the health of the ear canal has not been determined. We report that EDA-deficient mice, EDAR-deficient mice and EDARADD-deficient rats have Zymbal’s gland hypoplasia. EdaTa mice have 25% prevalence of otitis externa at postnatal day 21 and treatment with agonist anti-EDAR antibodies rescues Zymbal’s glands. The aetiopathogenesis of otitis externa involves infection with Gram-positive cocci, and dosing pregnant and lactating EdaTa females and pups with enrofloxacin reduces the prevalence of otitis externa. We infer that the deficit of sebum is the principal factor in predisposition to bacterial infection, and the EdaTa mouse is a potentially useful microbial challenge model for human acute otitis externa. Summary: Ectodysplasin-deficient mice have growth retardation of the auditory sebaceous Zymbal's gland and are predisposed to spontaneous bacterial infection of the outer ear canal by opportunistic pathogens.
Collapse
Affiliation(s)
- Jorge Del-Pozo
- Veterinary Pathology, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, Scotland, UK
| | - Denis J Headon
- Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, Scotland, UK
| | - James D Glover
- Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, Scotland, UK
| | - Ali Azar
- Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, Scotland, UK
| | | | - Mahmood F Bhutta
- Department of ENT, Royal Sussex County Hospital, Brighton BN2 5BE, UK.,Brighton and Sussex Medical School, Falmer Brighton BN1 9PX, UK
| | - Jon Riddell
- Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, Scotland, UK
| | - Scott Maxwell
- Veterinary Pathology, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, Scotland, UK
| | - Elspeth Milne
- Veterinary Pathology, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, Scotland, UK
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Michael Cheeseman
- Roslin Institute and The Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, Scotland, UK.,Division of Pathology, University of Edinburgh, Institute of Genetics & Molecular Medicine, Crewe Road, Edinburgh, EH4 2XR, Scotland, UK.,Centre for Comparative Pathology, Division of Pathology, University of Edinburgh, Institute of Genetics & Molecular Medicine, Crewe Road, Edinburgh, EH4 2XR, Scotland, UK
| |
Collapse
|
7
|
Albarnaz JD, Ren H, Torres AA, Shmeleva EV, Melo CA, Bannister AJ, Brember MP, Chung BYW, Smith GL. Molecular mimicry of NF-κB by vaccinia virus protein enables selective inhibition of antiviral responses. Nat Microbiol 2022; 7:154-168. [PMID: 34949827 PMCID: PMC7614822 DOI: 10.1038/s41564-021-01004-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 10/21/2021] [Indexed: 12/16/2022]
Abstract
Infection of mammalian cells with viruses activates NF-κB to induce the expression of cytokines and chemokines and initiate an antiviral response. Here, we show that a vaccinia virus protein mimics the transactivation domain of the p65 subunit of NF-κB to inhibit selectively the expression of NF-κB-regulated genes. Using co-immunoprecipitation assays, we found that the vaccinia virus protein F14 associates with NF-κB co-activator CREB-binding protein (CBP) and disrupts the interaction between p65 and CBP. This abrogates CBP-mediated acetylation of p65, after which it reduces promoter recruitment of the transcriptional regulator BRD4 and diminishes stimulation of NF-κB-regulated genes CXCL10 and CCL2. Recruitment of BRD4 to the promoters of NFKBIA and CXCL8 remains unaffected by either F14 or JQ1 (a competitive inhibitor of BRD4 bromodomains), indicating that BRD4 recruitment is acetylation-independent. Unlike other viral proteins that are general antagonists of NF-κB, F14 is a selective inhibitor of NF-κB-dependent gene expression. An in vivo model of infection demonstrated that F14 promotes virulence. Molecular mimicry of NF-κB may be conserved because other orthopoxviruses, including variola, monkeypox and cowpox viruses, encode orthologues of F14.
Collapse
Affiliation(s)
- Jonas D Albarnaz
- Department of Pathology, University of Cambridge, Cambridge, UK.
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| | - Hongwei Ren
- Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London, UK
| | - Alice A Torres
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Evgeniya V Shmeleva
- Department of Pathology, University of Cambridge, Cambridge, UK
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
| | - Carlos A Melo
- The Gurdon Institute, University of Cambridge, Cambridge, UK
| | | | | | - Betty Y-W Chung
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Geoffrey L Smith
- Department of Pathology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
8
|
Identification of loci associated with susceptibility to bovine paratuberculosis and with the dysregulation of the MECOM, eEF1A2, and U1 spliceosomal RNA expression. Sci Rep 2021; 11:313. [PMID: 33432064 PMCID: PMC7801378 DOI: 10.1038/s41598-020-79619-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Although genome-wide association studies have identified single nucleotide polymorphisms (SNPs) associated with the susceptibility to Mycobacterium avium subsp. paratuberculosis (MAP) infection, only a few functional mutations for bovine paratuberculosis (PTB) have been characterized. Expression quantitative trait loci (eQTLs) are genetic variants typically located in gene regulatory regions that alter gene expression in an allele-specific manner. eQTLs can be considered as functional links between genomic variants, gene expression, and ultimately phenotype. In the current study, peripheral blood (PB) and ileocecal valve (ICV) gene expression was quantified by RNA-Seq from fourteen Holstein cattle with no lesions and with PTB-associated histopathological lesions in gut tissues. Genotypes were generated from the Illumina LD EuroG10K BeadChip. The associations between gene expression levels (normalized read counts) and genetic variants were analyzed by a linear regression analysis using R Matrix eQTL 2.2. This approach allowed the identification of 192 and 48 cis-eQTLs associated with the expression of 145 and 43 genes in the PB and ICV samples, respectively. To investigate potential relationships between these cis-eQTLs and MAP infection, a case–control study was performed using the genotypes for all the identified cis-eQTLs and phenotypical data (histopathology, ELISA for MAP-antibodies detection, tissue PCR, and bacteriological culture) of 986 culled cows. Our results suggested that the heterozygous genotype in the cis-eQTL-rs43744169 (T/C) was associated with the up-regulation of the MDS1 and EVI1 complex (MECOM) expression, with positive ELISA, PCR, and bacteriological culture results, and with increased risk of progression to clinical PTB. As supporting evidence, the presence of the minor allele was associated with higher MECOM levels in plasma samples from infected cows and with increased MAP survival in an ex-vivo macrophage killing assay. Moreover, the presence of the two minor alleles in the cis-eQTL-rs110345285 (C/C) was associated with the dysregulation of the eukaryotic elongation factor 1-α2 (eEF1A2) expression and with increased ELISA (OD) values. Finally, the presence of the minor allele in the cis-eQTL rs109859270 (C/T) was associated with the up-regulation of the U1 spliceosomal RNA expression and with an increased risk of progression to clinical PTB. The introduction of these novel functional variants into marker-assisted breeding programs is expected to have a relevant effect on PTB control.
Collapse
|
9
|
Emerging Roles of PRDM Factors in Stem Cells and Neuronal System: Cofactor Dependent Regulation of PRDM3/16 and FOG1/2 (Novel PRDM Factors). Cells 2020; 9:cells9122603. [PMID: 33291744 PMCID: PMC7761934 DOI: 10.3390/cells9122603] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/13/2020] [Accepted: 11/25/2020] [Indexed: 12/19/2022] Open
Abstract
PRDI-BF1 (positive regulatory domain I-binding factor 1) and RIZ1 (retinoblastoma protein-interacting zinc finger gene 1) (PR) homologous domain containing (PRDM) transcription factors are expressed in neuronal and stem cell systems, and they exert multiple functions in a spatiotemporal manner. Therefore, it is believed that PRDM factors cooperate with a number of protein partners to regulate a critical set of genes required for maintenance of stem cell self-renewal and differentiation through genetic and epigenetic mechanisms. In this review, we summarize recent findings about the expression of PRDM factors and function in stem cell and neuronal systems with a focus on cofactor-dependent regulation of PRDM3/16 and FOG1/2. We put special attention on summarizing the effects of the PRDM proteins interaction with chromatin modulators (NuRD complex and CtBPs) on the stem cell characteristic and neuronal differentiation. Although PRDM factors are known to possess intrinsic enzyme activity, our literature analysis suggests that cofactor-dependent regulation of PRDM3/16 and FOG1/2 is also one of the important mechanisms to orchestrate bidirectional target gene regulation. Therefore, determining stem cell and neuronal-specific cofactors will help better understanding of PRDM3/16 and FOG1/2-controlled stem cell maintenance and neuronal differentiation. Finally, we discuss the clinical aspect of these PRDM factors in different diseases including cancer. Overall, this review will help further sharpen our knowledge of the function of the PRDM3/16 and FOG1/2 with hopes to open new research fields related to these factors in stem cell biology and neuroscience.
Collapse
|
10
|
EVI1 in Leukemia and Solid Tumors. Cancers (Basel) 2020; 12:cancers12092667. [PMID: 32962037 PMCID: PMC7564095 DOI: 10.3390/cancers12092667] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/02/2020] [Accepted: 09/13/2020] [Indexed: 11/30/2022] Open
Abstract
Simple Summary Ecotropic viral integration site 1 (EVI1) is transcriptionally activated in a subset of myeloid leukemias. Since its discovery, other isoforms of EVI1 have been identified. It has been shown that EVI1 and its isoforms mainly function as transcription factors and to play important roles not only in leukemia but also in a variety of solid tumors. To provide a comprehensive understanding of this family of proteins, we summarize the currently available knowledge of expression and function of EVI1 and its isoforms in leukemia and solid tumors and provide insights of future studies. Abstract The EVI1 gene encodes for a transcription factor with two zinc finger domains and is transcriptionally activated in a subset of myeloid leukemias. In leukemia, the transcriptional activation of EVI1 usually results from chromosomal rearrangements. Besides leukemia, EVI1 has also been linked to solid tumors including breast cancer, lung cancer, ovarian cancer and colon cancer. The MDS1/EVI1 gene is encoded by the same locus as EVI1. While EVI1 functions as a transcription repressor, MDS1/EVI1 acts as a transcription activator. The fusion protein encoded by the AML1/MDS1/EVI1 chimeric gene, resulting from chromosomal translocations in a subset of chronic myeloid leukemia, exhibits a similar function to EVI1. EVI1 has been shown to regulate cell proliferation, differentiation and apoptosis, whereas the functions of MDS1/EVI1 and AML1/MDS1/EVI1 remain elusive. In this review, we summarize the genetic structures, biochemical properties and biological functions of these proteins in cancer.
Collapse
|
11
|
Milne E, Nuttall T, Marioni-Henry K, Piccinelli C, Schwarz T, Azar A, Harris J, Duncan J, Cheeseman M. Cytological and microbiological characteristics of middle ear effusions in brachycephalic dogs. J Vet Intern Med 2020; 34:1454-1463. [PMID: 32407559 PMCID: PMC7379010 DOI: 10.1111/jvim.15792] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 12/20/2022] Open
Abstract
Background Middle ear effusion is common in brachycephalic dogs with similarities to otitis media with effusion in children. Association with the cranial and eustachian tube morphology and bacterial infection is suspected in both species. Hypothesis/objectives To determine cytological and bacteriological features of middle ear effusions in dogs, provide information on histological features, and further assess the dog as a model of the human disease. Animals Sixteen live dogs, 3 postmortem cases of middle ear effusion, and 2 postmortem controls. Methods Prospective; clinical investigation using computed tomography, magnetic resonance imaging, video‐otoscopy, myringotomy; cytological assessment of 30 and bacteriology of 28 effusions; histology and immunohistochemistry (CD3 for T‐lymphocytes, Pax5 for B lymphocytes and MAC387 for macrophages) of 10 middle ear sections. Results Effusions were associated with neurological deficits in 6/16 (38%) and concurrent atopic dermatitis and otitis externa in 9/16 (56%) of live cases. Neutrophils and macrophages predominated on cytology (median 60 [range 2%‐95.5%] and 27 [2%‐96.5%]) whether culture of effusions was positive or not. In histology sections, the mucosa was thickened in affected dogs but submucosal gland dilatation occurred in affected and unaffected dogs. There was no bacterial growth from 22/28 (79%) of effusions. Bacteria isolated from the other 6 (21%) were predominantly Staphylococcus pseudintermedius (4/6, 67%). Conclusions and Clinical Importance Clinical, morphological, and cytological findings in middle ear effusions of dogs and people suggest similar pathogeneses. Middle ear effusion of dogs could be a useful model of human otitis media with effusion. Such comparisons can improve understanding and management across species.
Collapse
Affiliation(s)
- Elspeth Milne
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Tim Nuttall
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Katia Marioni-Henry
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Chiara Piccinelli
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Tobias Schwarz
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Ali Azar
- MRC Centre for Regenerative Medicine, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, United Kingdom
| | - Jennifer Harris
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Juliet Duncan
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom
| | - Michael Cheeseman
- Royal (Dick) School of Veterinary Studies and The Roslin Institute, The University of Edinburgh, Midlothian, United Kingdom.,Centre for Comparative Pathology & Division of Pathology , University of Edinburgh, Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| |
Collapse
|
12
|
Geng R, Wang Q, Chen E, Zheng QY. Current Understanding of Host Genetics of Otitis Media. Front Genet 2020; 10:1395. [PMID: 32117425 PMCID: PMC7025460 DOI: 10.3389/fgene.2019.01395] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/20/2019] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of otitis media (OM), an inflammatory disease of the middle ear (ME), involves interplay between many different factors, including the pathogenicity of infectious pathogens, host immunological status, environmental factors, and genetic predisposition, which is known to be a key determinant of OM susceptibility. Animal models and human genetics studies have identified many genes and gene variants associated with OM susceptibility: genes that encode components of multiple signaling pathways involved in host immunity and inflammatory responses of the ME mucosa; genes involved in cellular function, such as mucociliary transport, mucin production, and mucous cell metaplasia; and genes that are essential for Eustachian tube (ET) development, ME cavitation, and homeostasis. Since our last review, several new mouse models with mutations in genes such as CCL3, IL-17A, and Nisch have been reported. Moreover, genetic variants and polymorphisms in several genes, including FNDC1, FUT2, A2ML1, TGIF1, CD44, and IL1-RA variable number tandem repeat (VNTR) allele 2, have been identified as being significantly associated with OM. In this review, we focus on the current understanding of the role of host genetics in OM, including recent discoveries and future research prospects. Further studies on the genes identified thus far and the discovery of new genes using advanced technologies such as gene editing, next generation sequencing, and genome-wide association studies, will advance our understanding of the molecular mechanism underlying the pathogenesis of OM and provide new avenues for early screening and developing effective preventative and therapeutic strategies to treat OM.
Collapse
Affiliation(s)
- Ruishuang Geng
- College of Special Education, Binzhou Medical University, Yantai, China
| | - Qingzhu Wang
- College of Special Education, Binzhou Medical University, Yantai, China.,Department of Otolaryngology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Eileen Chen
- Department of Otolaryngology, Case Western Reserve University, Cleveland, OH, United States
| | - Qing Yin Zheng
- Department of Otolaryngology, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
13
|
Vikhe PP, Purnell T, Brown SDM, Hood DW. Cellular content plays a crucial role in Non-typeable Haemophilus influenzae infection of preinflamed Junbo mouse middle ear. Cell Microbiol 2018; 21:e12960. [PMID: 30265765 PMCID: PMC6491974 DOI: 10.1111/cmi.12960] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/19/2018] [Accepted: 09/24/2018] [Indexed: 01/12/2023]
Abstract
Non-typeable Haemophilus influenzae (NTHi) is a major pathogen causing acute otitis media (AOM). The relationship between the cellular content of the middle ear fluid (MEF) during AOM and infection of NTHi is poorly understood. Using the Junbo mouse, a characterised NTHi infection model, we analysed the cellular content of MEF and correlated the data with NTHi titres. The MEF of the Junbo mouse was heterogeneous between ears and was graded from 1 to 5; 1 being highly serous/clear and 5 being heavily viscous/opaque. At seven-day post-intranasal inoculation, NTHi was not found in grade-1 or 2 fluids, and the proportion of MEF that supported NTHi increased with the grade. Analyses by flow cytometry indicated that the cellular content was highest in grade-4 and 5 fluids, with a greater proportion of necrotic cells and a low-live cell count. NTHi infection of the middle ear increased the cell count and led to infiltration of immune cells and changes in the cytokine and chemokine levels. Following NTHi inoculation, high-grade infected MEFs had greater neutrophil infiltration whereas monocyte infiltration was significantly higher in serous noninfected low-grade fluids. These data underline a role for immune cells, specifically monocytes and neutrophils, and cell necrosis in NTHi infection of the Junbo mouse middle ear.
Collapse
Affiliation(s)
- Pratik P Vikhe
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Tom Purnell
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Steve D M Brown
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Derek W Hood
- Mammalian Genetics Unit, MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| |
Collapse
|
14
|
EVI1 overexpression reprograms hematopoiesis via upregulation of Spi1 transcription. Nat Commun 2018; 9:4239. [PMID: 30315161 PMCID: PMC6185954 DOI: 10.1038/s41467-018-06208-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 08/21/2018] [Indexed: 01/19/2023] Open
Abstract
Inv(3q26) and t(3:3)(q21;q26) are specific to poor-prognosis myeloid malignancies, and result in marked overexpression of EVI1, a zinc-finger transcription factor and myeloid-specific oncoprotein. Despite extensive study, the mechanism by which EVI1 contributes to myeloid malignancy remains unclear. Here we describe a new mouse model that mimics the transcriptional effects of 3q26 rearrangement. We show that EVI1 overexpression causes global distortion of hematopoiesis, with suppression of erythropoiesis and lymphopoiesis, and marked premalignant expansion of myelopoiesis that eventually results in leukemic transformation. We show that myeloid skewing is dependent on DNA binding by EVI1, which upregulates Spi1, encoding master myeloid regulator PU.1. We show that EVI1 binds to the -14 kb upstream regulatory element (-14kbURE) at Spi1; knockdown of Spi1 dampens the myeloid skewing. Furthermore, deletion of the -14kbURE at Spi1 abrogates the effects of EVI1 on hematopoietic stem cells. These findings support a novel mechanism of leukemogenesis through EVI1 overexpression.
Collapse
|
15
|
Bhutta MF, Thornton RB, Kirkham LAS, Kerschner JE, Cheeseman MT. Understanding the aetiology and resolution of chronic otitis media from animal and human studies. Dis Model Mech 2018; 10:1289-1300. [PMID: 29125825 PMCID: PMC5719252 DOI: 10.1242/dmm.029983] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inflammation of the middle ear, known clinically as chronic otitis media, presents in different forms, such as chronic otitis media with effusion (COME; glue ear) and chronic suppurative otitis media (CSOM). These are highly prevalent diseases, especially in childhood, and lead to significant morbidity worldwide. However, much remains unclear about this disease, including its aetiology, initiation and perpetuation, and the relative roles of mucosal and leukocyte biology, pathogens, and Eustachian tube function. Chronic otitis media is commonly modelled in mice but most existing models only partially mimic human disease and many are syndromic. Nevertheless, these models have provided insights into potential disease mechanisms, and have implicated altered immune signalling, mucociliary function and Eustachian tube function as potential predisposing mechanisms. Clinical studies of chronic otitis media have yet to implicate a particular molecular pathway or mechanism, and current human genetic studies are underpowered. We also do not fully understand how existing interventions, such as tympanic membrane repair, work, nor how chronic otitis media spontaneously resolves. This Clinical Puzzle article describes our current knowledge of chronic otitis media and the existing research models for this condition. It also identifies unanswered questions about its pathogenesis and treatment, with the goal of advancing our understanding of this disease to aid the development of novel therapeutic interventions. Summary: Chronic middle ear inflammation is a common disease. Animal models, and in particular mouse models, have been used to elucidate some potential mechanisms, including dysfunction in immune signalling, mucociliary function or Eustachian tube function.
Collapse
Affiliation(s)
- Mahmood F Bhutta
- Department of ENT, Brighton and Sussex University Hospitals NHS Trust, Brighton, BN2 5BE, England .,Division of Paediatrics, University of Western Australia, Subiaco, WA 6008, Australia
| | - Ruth B Thornton
- Division of Paediatrics, University of Western Australia, Subiaco, WA 6008, Australia.,Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Subiaco, WA 6008, Australia
| | - Lea-Ann S Kirkham
- Division of Paediatrics, University of Western Australia, Subiaco, WA 6008, Australia.,Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, Subiaco, WA 6008, Australia
| | - Joseph E Kerschner
- Office of the Dean, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael T Cheeseman
- Division of Developmental Biology, Roslin Institute, University of Edinburgh, Midlothian, EH23 9RG, Scotland
| |
Collapse
|
16
|
Mulay A, Hood DW, Williams D, Russell C, Brown SDM, Bingle L, Cheeseman M, Bingle CD. Loss of the homeostatic protein BPIFA1, leads to exacerbation of otitis media severity in the Junbo mouse model. Sci Rep 2018; 8:3128. [PMID: 29449589 PMCID: PMC5814562 DOI: 10.1038/s41598-018-21166-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/30/2018] [Indexed: 02/02/2023] Open
Abstract
Otitis Media (OM) is characterized by epithelial abnormalities and defects in innate immunity in the middle ear (ME). Although, BPIFA1, a member of the BPI fold containing family of putative innate defence proteins is abundantly expressed by the ME epithelium and SNPs in Bpifa1 have been associated with OM susceptibility, its role in the ME is not well characterized. We investigated the role of BPIFA1 in protection of the ME and the development of OM using murine models. Loss of Bpifa1 did not lead to OM development. However, deletion of Bpifa1 in Evi1Jbo/+ mice, a model of chronic OM, caused significant exacerbation of OM severity, thickening of the ME mucosa and increased collagen deposition, without a significant increase in pro-inflammatory gene expression. Our data suggests that BPIFA1 is involved in maintaining homeostasis within the ME under steady state conditions and its loss in the presence of inflammation, exacerbates epithelial remodelling leading to more severe OM.
Collapse
Affiliation(s)
- Apoorva Mulay
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Derek W Hood
- MRC Mammalian Genetics Unit, MRC Harwell Institute, Didcot, UK
| | - Debbie Williams
- MRC Mammalian Genetics Unit, MRC Harwell Institute, Didcot, UK
| | - Catherine Russell
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Steve D M Brown
- MRC Mammalian Genetics Unit, MRC Harwell Institute, Didcot, UK
| | - Lynne Bingle
- Oral and Maxillofacial Pathology, Department of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Michael Cheeseman
- Roslin Institute, University of Edinburgh, Edinburgh, UK.,Division of Pathology, University of Edinburgh, Edinburgh, UK
| | - Colin D Bingle
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK. .,Florey Institute for Host Pathogen Interactions, University of Sheffield, Sheffield, UK.
| |
Collapse
|
17
|
Bioenergetic state regulates innate inflammatory responses through the transcriptional co-repressor CtBP. Nat Commun 2017; 8:624. [PMID: 28935892 PMCID: PMC5608947 DOI: 10.1038/s41467-017-00707-0] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2014] [Accepted: 07/21/2017] [Indexed: 01/04/2023] Open
Abstract
The innate inflammatory response contributes to secondary injury in brain trauma and other disorders. Metabolic factors such as caloric restriction, ketogenic diet, and hyperglycemia influence the inflammatory response, but how this occurs is unclear. Here, we show that glucose metabolism regulates pro-inflammatory NF-κB transcriptional activity through effects on the cytosolic NADH:NAD+ ratio and the NAD(H) sensitive transcriptional co-repressor CtBP. Reduced glucose availability reduces the NADH:NAD+ ratio, NF-κB transcriptional activity, and pro-inflammatory gene expression in macrophages and microglia. These effects are inhibited by forced elevation of NADH, reduced expression of CtBP, or transfection with an NAD(H) insensitive CtBP, and are replicated by a synthetic peptide that inhibits CtBP dimerization. Changes in the NADH:NAD+ ratio regulate CtBP binding to the acetyltransferase p300, and regulate binding of p300 and the transcription factor NF-κB to pro-inflammatory gene promoters. These findings identify a mechanism by which alterations in cellular glucose metabolism can influence cellular inflammatory responses. Several metabolic factors affect cellular glucose metabolism as well as the innate inflammatory response. Here, the authors show that glucose metabolism regulates pro-inflammatory responses through effects on the cytosolic NADH:NAD+ ratio and the NAD(H)-sensitive transcription co-repressor CtBP.
Collapse
|
18
|
A mutation in Nischarin causes otitis media via LIMK1 and NF-κB pathways. PLoS Genet 2017; 13:e1006969. [PMID: 28806779 PMCID: PMC5570507 DOI: 10.1371/journal.pgen.1006969] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/24/2017] [Accepted: 08/08/2017] [Indexed: 01/18/2023] Open
Abstract
Otitis media (OM), inflammation of the middle ear (ME), is a common cause of conductive hearing impairment. Despite the importance of the disease, the aetiology of chronic and recurrent forms of middle ear inflammatory disease remains poorly understood. Studies of the human population suggest that there is a significant genetic component predisposing to the development of chronic OM, although the underlying genes are largely unknown. Using N-ethyl-N-nitrosourea mutagenesis we identified a recessive mouse mutant, edison, that spontaneously develops a conductive hearing loss due to chronic OM. The causal mutation was identified as a missense change, L972P, in the Nischarin (NISCH) gene. edison mice develop a serous or granulocytic effusion, increasingly macrophage and neutrophil rich with age, along with a thickened, inflamed mucoperiosteum. We also identified a second hypomorphic allele, V33A, with only modest increases in auditory thresholds and reduced incidence of OM. NISCH interacts with several proteins, including ITGA5 that is thought to have a role in modulating VEGF-induced angiogenesis and vascularization. We identified a significant genetic interaction between Nisch and Itga5; mice heterozygous for Itga5-null and homozygous for edison mutations display a significantly increased penetrance and severity of chronic OM. In order to understand the pathological mechanisms underlying the OM phenotype, we studied interacting partners to NISCH along with downstream signalling molecules in the middle ear epithelia of edison mouse. Our analysis implicates PAK1 and RAC1, and downstream signalling in LIMK1 and NF-κB pathways in the development of chronic OM. Otitis media (OM) is the most common cause of deafness in children and is primarily characterised by inflammation of the middle ear. It is the most common cause of surgery in children in the developed world, with many children developing recurrent and chronic forms of OM undergoing tympanostomy tube insertion. There is evidence that a significant genetic component contributes towards the development of recurrent and chronic forms of OM. The mouse has been a powerful tool for identifying the genes involved in chronic OM. In this study we identified and characterised edison, a novel mouse model of chronic OM that shares important features with the chronic disease in humans. A mutation in the Nisch gene causes edison mice to spontaneously develop OM following birth and subsequently develop chronic OM, with an associated hearing loss. Our molecular analysis of the mutation reveals the underlying pathological mechanisms and pathways involved in OM in the edison mouse, involving PAK1, RAC1 and downstream signalling in LIMK1 and NF-κB pathways. Identification of the edison mutant provides an important genetic disease model of chronic OM and implicates a new gene and genetic pathways involved in predisposition to OM.
Collapse
|
19
|
Colombo AR, Zubair A, Thiagarajan D, Nuzhdin S, Triche TJ, Ramsingh G. Suppression of Transposable Elements in Leukemic Stem Cells. Sci Rep 2017; 7:7029. [PMID: 28765607 PMCID: PMC5539300 DOI: 10.1038/s41598-017-07356-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/23/2017] [Indexed: 01/15/2023] Open
Abstract
Genomic transposable elements (TEs) comprise nearly half of the human genome. The expression of TEs is considered potentially hazardous, as it can lead to insertional mutagenesis and genomic instability. However, recent studies have revealed that TEs are involved in immune-mediated cell clearance. Hypomethylating agents can increase the expression of TEs in cancer cells, inducing ‘viral mimicry’, causing interferon signalling and cancer cell killing. To investigate the role of TEs in the pathogenesis of acute myeloid leukaemia (AML), we studied TE expression in several cell fractions of AML while tracking its development (pre-leukemic haematopoietic stem cells, leukemic stem cells [LSCs], and leukemic blasts). LSCs, which are resistant to chemotherapy and serve as reservoirs for relapse, showed significant suppression of TEs and interferon pathways. Similarly, high-risk cases of myelodysplastic syndrome (MDS) showed far greater suppression of TEs than low-risk cases. We propose TE suppression as a mechanism for immune escape in AML and MDS. Repression of TEs co-occurred with the upregulation of several genes known to modulate TE expression, such as RNA helicases and autophagy genes. Thus, we have identified potential pathways that can be targeted to activate cancer immunogenicity via TEs in AML and MDS.
Collapse
Affiliation(s)
- Anthony R Colombo
- Keck School of Medicine of University of Southern California, Jane Anne Nohl Division of Hematology and Center for the Study of Blood Diseases, Los Angeles, California, 90033, USA
| | - Asif Zubair
- University of Southern California, Department of Molecular and Computational Biology, Los Angeles, CA, 90089-2910, USA
| | - Devi Thiagarajan
- Keck School of Medicine of University of Southern California, Jane Anne Nohl Division of Hematology and Center for the Study of Blood Diseases, Los Angeles, California, 90033, USA.,Langone Medical Center of New York University School of Medicine, Endocrinology Division for the Study of Diabetes, 550 1st Avenue, New York, NY, 10016, USA
| | - Sergey Nuzhdin
- University of Southern California, Department of Molecular and Computational Biology, Los Angeles, CA, 90089-2910, USA
| | - Timothy J Triche
- Keck School of Medicine of University of Southern California, Jane Anne Nohl Division of Hematology and Center for the Study of Blood Diseases, Los Angeles, California, 90033, USA.
| | - Giridharan Ramsingh
- Keck School of Medicine of University of Southern California, Jane Anne Nohl Division of Hematology and Center for the Study of Blood Diseases, Los Angeles, California, 90033, USA.
| |
Collapse
|
20
|
Abstract
Acute otitis media, inflammation of the middle ear bulla, is the most common bacterial infection in children. For one of the principal otopathogens, non-typeable Haemophilus influenzae (NTHi), animal models allow us to investigate host-microbial interactions relevant to the onset and progression of infection and to study treatment of middle ear disease. We have established a robust model of NTHi middle ear infection in the Junbo mouse. Intranasal inoculation with NTHi produces high rates of bulla infection and high bacterial titers in bulla fluids; bacteria can also spread down the respiratory tract to the mouse lung. An innate immune response is detected in the bulla of Junbo mice following NTHi infection, and bacteria are maintained in some ears at least up to day 56 post-inoculation. The Junbo/NTHi infection model facilitates studies on bacterial pathogenesis and antimicrobial intervention regimens and vaccines for better treatment and prevention of NTHi middle ear infection. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Michael T Cheeseman
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies University of Edinburgh, Easter Bush, United Kingdom
| | - Derek W Hood
- Molecular Genetics Unit, MRC Harwell Institute, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom
| |
Collapse
|
21
|
Yang CJ, Liu YP, Dai HY, Shiue YL, Tsai CJ, Huang MS, Yeh YT. Nuclear HDAC6 inhibits invasion by suppressing NF-κB/MMP2 and is inversely correlated with metastasis of non-small cell lung cancer. Oncotarget 2016; 6:30263-76. [PMID: 26388610 PMCID: PMC4745796 DOI: 10.18632/oncotarget.4749] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 09/04/2015] [Indexed: 11/25/2022] Open
Abstract
Histone deacetylase 6 (HDAC6) is a unique member of the histone deacetylase family. Although HDAC6 is mainly localized in the cytoplasm, it can regulate the activities of the transcription factors in the nucleus. However, a correlation of intracellular distribution of HDAC6 with tumor progression is lacking. In this study, we found that a low frequency of nuclear HDAC6-positive cells in tumors was associated with distant metastasis and a worse overall survival in 134 patients with non-small cell lung cancer (NSCLC). Ectopic expression of wild-type HDAC6 promoted migration and invasion of A549 and H661 cells. However, the enforced expression of nuclear export signal-deleted HDAC6 inhibited the invasion but not the migration of both cell lines. The inhibitory effect of nuclear HDAC6 on invasion was mediated by the deacetylation of the p65 subunit of nuclear factor-κB, which decreased its DNA-binding activity to the MMP2 promoter, leading to the downregulation of MMP2 expression. Our findings indicated that the loss of nuclear HDAC6 may be a potential biomarker for predicting metastasis in patients with NSCLC.
Collapse
Affiliation(s)
- Chih-Jen Yang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Peng Liu
- Department of Genome Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hong-Ying Dai
- Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chia-Jung Tsai
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ming-Shyan Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Tsung Yeh
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| |
Collapse
|
22
|
Azar A, Piccinelli C, Brown H, Headon D, Cheeseman M. Ectodysplasin signalling deficiency in mouse models of hypohidrotic ectodermal dysplasia leads to middle ear and nasal pathology. Hum Mol Genet 2016; 25:3564-3577. [PMID: 27378689 PMCID: PMC5179950 DOI: 10.1093/hmg/ddw202] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/21/2016] [Accepted: 06/21/2016] [Indexed: 12/14/2022] Open
Abstract
Hypohidrotic ectodermal dysplasia (HED) results from mutation of the EDA, EDAR or EDARADD genes and is characterized by reduced or absent eccrine sweat glands, hair follicles and teeth, and defective formation of salivary, mammary and craniofacial glands. Mouse models with HED also carry Eda, Edar or Edaradd mutations and have defects that map to the same structures. Patients with HED have ear, nose and throat disease, but this has not been investigated in mice bearing comparable genetic mutations. We report that otitis media, rhinitis and nasopharyngitis occur at high frequency in Eda and Edar mutant mice and explore the pathogenic mechanisms related to glandular function, microbial and immune parameters in these lines. Nasopharynx auditory tube glands fail to develop in HED mutant mice and the functional implications include loss of lysozyme secretion, reduced mucociliary clearance and overgrowth of nasal commensal bacteria accompanied by neutrophil exudation. Heavy nasopharynx foreign body load and loss of gland protection alters the auditory tube gating function and the auditory tubes can become pathologically dilated. Accumulation of large foreign body particles in the bulla stimulates granuloma formation. Analysis of immune cell populations and myeloid cell function shows no evidence of overt immune deficiency in HED mutant mice. Our findings using HED mutant mice as a model for the human condition support the idea that ear and nose pathology in HED patients arises as a result of nasal and nasopharyngeal gland deficits, reduced mucociliary clearance and impaired auditory tube gating function underlies the pathological sequelae in the bulla.
Collapse
Affiliation(s)
| | - Chiara Piccinelli
- Veterinary Pathology, The Royal (Dick) School of Veterinary Studies, University of Edinburgh, EH25 9RG, Scotland, UK
| | - Helen Brown
- Genetics and Genomics Division, Roslin Institute and The Royal (Dick) School of Veterinary Studies
| | | | | |
Collapse
|
23
|
Zhang J, Zhang Y, Xiao F, Liu Y, Wang J, Gao H, Rong S, Yao Y, Li J, Xu G. The peroxisome proliferator-activated receptor γ agonist pioglitazone prevents NF-κB activation in cisplatin nephrotoxicity through the reduction of p65 acetylation via the AMPK-SIRT1/p300 pathway. Biochem Pharmacol 2015; 101:100-11. [PMID: 26673543 DOI: 10.1016/j.bcp.2015.11.027] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/30/2015] [Indexed: 01/11/2023]
Abstract
The thiazolidinedione pioglitazone, which is also a PPAR-γ agonist, now is widely used in patients with hypercholesterolemia and hypertriglyceridemia. NF-κB is a ubiquitously expressed transcription factor controlling the expression of numerous genes involved in inflammation. The aim of the present study was to evaluate whether the activation of PPAR-γ attenuates the cisplatin-induced NF-κB activation in cisplatin nephrotoxicity. The results showed that the PPAR-γ agonist pioglitazone decreased the expression of NF-κB p65 transcription target genes (e.g., IL-6, IL-1β, and TNF-α) and inhibited histological injury and inflammatory cells infiltration in cisplatin nephrotoxicity. The suppression of NF-κB activity following pioglitazone treatment inhibited the cisplatin-induced IκB-α degredation and NF-κB p65 subunit translocation. Translocation of the NF-κB p65 subunit depends on p65 acetylation, which primarily regulated by SIRT1 or p300. Notably, AMP kinase (AMPK) activation not only decreased the phosphorylation, activation and p65 interaction of p300 but also increased SIRT1 expression, activation and p65 binding, thus leading to a significant reduction in p65 acetylation. Interestingly, the reduction of IL-6, TNF-α and IL-1β, the inhibition of histological injury and the inflammatory cells infiltration following pioglitazone treatment in cisplatin nephrotoxicity were attenuated after treatment with the PPAR-γ antagonist GW9662. These results suggest that the PPAR-γ agonist pioglitazone prevents NF-κB activation in cisplatin nephrotoxicity through a reduction in p65 acetylation via the AMPK-SIRT1/p300 pathway.
Collapse
Affiliation(s)
- Jiong Zhang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Department of Nephrology, University of Electronic Science and Technology, Sichuan Academy of Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Ying Zhang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Fang Xiao
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong Unversity of Science and Technology, China
| | - Yanyan Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jin Wang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Hongyu Gao
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Song Rong
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Ying Yao
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Junhua Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
24
|
Hood D, Moxon R, Purnell T, Richter C, Williams D, Azar A, Crompton M, Wells S, Fray M, Brown SDM, Cheeseman MT. A new model for non-typeable Haemophilus influenzae middle ear infection in the Junbo mutant mouse. Dis Model Mech 2015; 9:69-79. [PMID: 26611891 PMCID: PMC4728332 DOI: 10.1242/dmm.021659] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 11/15/2015] [Indexed: 01/23/2023] Open
Abstract
Acute otitis media, inflammation of the middle ear, is the most common bacterial infection in children and, as a consequence, is the most common reason for antimicrobial prescription to this age group. There is currently no effective vaccine for the principal pathogen involved, non-typeable Haemophilus influenzae (NTHi). The most frequently used and widely accepted experimental animal model of middle ear infection is in chinchillas, but mice and gerbils have also been used. We have established a robust model of middle ear infection by NTHi in the Junbo mouse, a mutant mouse line that spontaneously develops chronic middle ear inflammation in specific pathogen-free conditions. The heterozygote Junbo mouse (Jbo/+) bears a mutation in a gene (Evi1, also known as Mecom) that plays a role in host innate immune regulation; pre-existing middle ear inflammation promotes NTHi middle ear infection. A single intranasal inoculation with NTHi produces high rates (up to 90%) of middle ear infection and bacterial titres (104-105 colony-forming units/µl) in bulla fluids. Bacteria are cleared from the majority of middle ears between day 21 and 35 post-inoculation but remain in approximately 20% of middle ears at least up to day 56 post-infection. The expression of Toll-like receptor-dependent response cytokine genes is elevated in the middle ear of the Jbo/+ mouse following NTHi infection. The translational potential of the Junbo model for studying antimicrobial intervention regimens was shown using a 3 day course of azithromycin to clear NTHi infection, and its potential use in vaccine development studies was shown by demonstrating protection in mice immunized with killed homologous, but not heterologous, NTHi bacteria. Summary: Acute otitis media is an important disease in children. We describe a new infection model for translational research that uses the Junbo mouse mutant intranasally inoculated with non-typeable Haemophilus influenzae.
Collapse
Affiliation(s)
- Derek Hood
- MRC Mammalian Genetics Unit, MRC Harwell, Didcot, Oxford, OX11 0RD, UK
| | - Richard Moxon
- Department of Paediatrics, University of Oxford Medical Sciences Division, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Tom Purnell
- MRC Mammalian Genetics Unit, MRC Harwell, Didcot, Oxford, OX11 0RD, UK
| | - Caroline Richter
- MRC Mammalian Genetics Unit, MRC Harwell, Didcot, Oxford, OX11 0RD, UK
| | - Debbie Williams
- MRC Mammalian Genetics Unit, MRC Harwell, Didcot, Oxford, OX11 0RD, UK
| | - Ali Azar
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, Easter Bush Campus, University of Edinburgh, EH25 9RG, UK
| | - Michael Crompton
- MRC Mammalian Genetics Unit, MRC Harwell, Didcot, Oxford, OX11 0RD, UK
| | - Sara Wells
- Mary Lyon Centre, MRC Harwell, Harwell, Didcot, Oxford, OX11 0RD, UK
| | - Martin Fray
- Mary Lyon Centre, MRC Harwell, Harwell, Didcot, Oxford, OX11 0RD, UK
| | - Steve D M Brown
- MRC Mammalian Genetics Unit, MRC Harwell, Didcot, Oxford, OX11 0RD, UK
| | - Michael T Cheeseman
- MRC Mammalian Genetics Unit, MRC Harwell, Didcot, Oxford, OX11 0RD, UK Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, Easter Bush Campus, University of Edinburgh, EH25 9RG, UK Mary Lyon Centre, MRC Harwell, Harwell, Didcot, Oxford, OX11 0RD, UK
| |
Collapse
|
25
|
Su YC, Mukherjee O, Singh B, Hallgren O, Westergren-Thorsson G, Hood D, Riesbeck K. Haemophilus influenzae P4 Interacts With Extracellular Matrix Proteins Promoting Adhesion and Serum Resistance. J Infect Dis 2015; 213:314-23. [PMID: 26153407 DOI: 10.1093/infdis/jiv374] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/26/2015] [Indexed: 02/07/2023] Open
Abstract
Interaction with the extracellular matrix (ECM) is one of the successful colonization strategies employed by nontypeable Haemophilus influenzae (NTHi). Here we identified Haemophilus lipoprotein e (P4) as a receptor for ECM proteins. Purified recombinant P4 displayed a high binding affinity for laminin (Kd = 9.26 nM) and fibronectin (Kd = 10.19 nM), but slightly less to vitronectin (Kd = 16.51 nM). A P4-deficient NTHi mutant showed a significantly decreased binding to these ECM components. Vitronectin acquisition conferred serum resistance to both P4-expressing NTHi and Escherichia coli transformants. P4-mediated bacterial adherence to pharynx, type II alveolar, and bronchial epithelial cells was mainly attributed to fibronectin. Importantly, a significantly reduced bacterial infection was observed in the middle ear of the Junbo mouse model when NTHi was devoid of P4. In conclusion, our data provide new insight into the role of P4 as an important factor for Haemophilus colonization and subsequent respiratory tract infection.
Collapse
Affiliation(s)
- Yu-Ching Su
- Clinical Microbiology, Department of Translational Medicine, Lund University, Malmö
| | - Oindrilla Mukherjee
- Clinical Microbiology, Department of Translational Medicine, Lund University, Malmö
| | - Birendra Singh
- Clinical Microbiology, Department of Translational Medicine, Lund University, Malmö
| | - Oskar Hallgren
- Department for Experimental Medical Sciences Department of Respiratory Medicine and Allergology, Lund University, Sweden
| | | | - Derek Hood
- Mammalian Genetics Unit, MRC Harwell, Harwell Science & Innovation Campus, Oxfordshire, United Kingdom
| | - Kristian Riesbeck
- Clinical Microbiology, Department of Translational Medicine, Lund University, Malmö
| |
Collapse
|
26
|
Bhutta MF, Cheeseman MT, Brown SDM. Myringotomy in the Junbo mouse model of chronic otitis media alleviates inflammation and cellular hypoxia. Laryngoscope 2014; 124:E377-83. [PMID: 24706577 DOI: 10.1002/lary.24698] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/14/2014] [Accepted: 04/01/2014] [Indexed: 01/09/2023]
Abstract
OBJECTIVES/HYPOTHESIS Ventilation of the chronically inflamed middle ear is a key outcome in functional middle ear surgery. Grommets eliminate middle ear effusion, but there is also evidence that they downregulate inflammation. The reason for this is not understood, but there is little to suggest alteration in eustachian tube ventilatory capacity. Previous work has shown that the Junbo mouse model of chronic otitis media has hypoxic middle ear mucosa and bulla fluid leucocytes. Here we explore whether surgical ventilation may alleviate chronic otitis media through downregulation of hypoxia. STUDY DESIGN Surgical intervention on a mouse model of disease. METHODS We established patency of myringotomy incision as 5 days in wild-type mice. We performed unilateral myringotomy on three cohorts of mice: 10 wild-type controls, 12 Junbo mice, and 15 Junbo mice with additional removal of middle ear effusion. A small cohort of these mice were labeled in vivo by intraperitoneal injection of pimonidazole to identify tissue hypoxia. Tissues were assessed for mucoperiosteal thickening and pimonidazole labeling, comparing operated to nonoperated ears. RESULTS Ventilation of the inflamed Junbo middle ear revealed significant reduction in inflammatory thickening associated with loss of pimonidazole labeling, suggesting resolution of cellular hypoxia. CONCLUSIONS Surgical ventilation may achieve therapeutic effect through alleviation of cellular hypoxia in the chronically inflamed middle ear. Targeted molecular therapy of hypoxia signaling may offer future alternative therapy for chronic otitis media.
Collapse
Affiliation(s)
- Mahmood F Bhutta
- UCL Ear Institute, London, United Kingdom; MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, United Kingdom; Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom; Department of Otolaryngology, Barts Health NHS Trust, London, United Kingdom
| | | | | |
Collapse
|
27
|
Sun H, Wu Y, Fu D, Liu Y, Huang C. SIRT6 Regulates Osteogenic Differentiation of Rat Bone Marrow Mesenchymal Stem Cells Partially via Suppressing the Nuclear Factor-κB Signaling Pathway. Stem Cells 2014; 32:1943-55. [DOI: 10.1002/stem.1671] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 01/19/2014] [Accepted: 01/26/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Hualing Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei People's Republic of China
| | - Yanru Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei People's Republic of China
| | - Dongjie Fu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei People's Republic of China
| | - Yinchen Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei People's Republic of China
| | - Cui Huang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine Ministry of Education; School and Hospital of Stomatology; Wuhan University; Wuhan Hubei People's Republic of China
| |
Collapse
|
28
|
Porcine reproductive and respiratory syndrome virus induces IL-1β production depending on TLR4/MyD88 pathway and NLRP3 inflammasome in primary porcine alveolar macrophages. Mediators Inflamm 2014; 2014:403515. [PMID: 24966466 PMCID: PMC4055429 DOI: 10.1155/2014/403515] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/23/2014] [Accepted: 04/23/2014] [Indexed: 12/17/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is an Arterivirus that has been devastating the swine industry worldwide since the late 1980s. Previous studies have reported that PRRSV infection induced the production of IL-1β. However, the cellular sensors and signaling pathways involved in this process have not been elucidated yet. Here, we studied the mechanisms responsible for the production of IL-1β in response to highly pathogenic PRRSV. Upon PRRSV infection of primary porcine alveolar macrophages, both mRNA expression and secretion of IL-1β were significantly increased in a time- and dose-dependent manner. We also investigated the role of several pattern-recognition receptors and adaptor molecules in this response and showed that the TLR4/MyD88 pathway and its downstream signaling molecules, NF-κB, ERK1/2, and p38 MAPKs, were involved in IL-1β production during PRRSV infection. Treatment with specific inhibitors or siRNA knockdown assays demonstrated that components of the NLRP3 inflammasome were crucial for IL-1β secretion but not for IL-1β mRNA expression. Furthermore, TLR4/MyD88/NF-κB signaling pathway was involved in PRRSV-induced expression of NLRP3 inflammasome components. Together, our results deciphered the pathways leading from recognition of PRRSV to the production and release of IL-1β, providing a deeper knowledge of the mechanisms of PRRSV-induced inflammation responses.
Collapse
|
29
|
Abstract
Otitis media (OM) is a common cause of childhood hearing loss. The large medical costs involved in treating this condition have meant that research to understand the pathology of this disease and identify new therapeutic interventions is important. There is evidence that susceptibility to OM has a significant genetic component, although little is known about the key genetic pathways involved. Mouse models for disease have become an important resource to understand a variety of human pathologies, including OM, due to the ability to easily manipulate their genetic components. This has enabled researchers to create models of acute OM, and has aided in the identification of a number of new genes associated with chronic disease, through the use of mutagenesis programs. The use of mouse models has identified a number of key molecular signalling pathways involved in the development of this condition, with genes identified from models shown to be associated with human OM.
Collapse
|
30
|
Komatsu K, Lee JY, Miyata M, Hyang Lim J, Jono H, Koga T, Xu H, Yan C, Kai H, Li JD. Inhibition of PDE4B suppresses inflammation by increasing expression of the deubiquitinase CYLD. Nat Commun 2013; 4:1684. [PMID: 23575688 PMCID: PMC3644066 DOI: 10.1038/ncomms2674] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Accepted: 02/27/2013] [Indexed: 12/28/2022] Open
Abstract
The deubiquitinase CYLD acts as a key negative regulator to tightly control overactive inflammation. Most anti-inflammatory strategies have focused on directly targeting the positive regulator, which often results in significant side effects such as suppression of the host defence response. Here, we show that inhibition of phosphodiesterase 4B (PDE4B) markedly enhances upregulation of CYLD expression in response to bacteria, thereby suggesting that PDE4B acts as a negative regulator for CYLD. Interestingly, in Cyld-deficient mice, inhibition of PDE4B no longer suppresses inflammation. Moreover, PDE4B negatively regulates CYLD via specific activation of JNK2 but not JNK1. Importantly, ototopical post-inoculation administration of a PDE4 inhibitor suppresses inflammation in this animal model, thus demonstrating the therapeutic potential of targeting PDE4. These studies provide insights into how inflammation is tightly regulated via the inhibition of its negative regulator and may also lead to the development of new anti-inflammatory therapeutics that upregulate CYLD expression. Phosphodiesterase 4 inhibitors are under development as anti-inflammatory drugs, however, their mechanisms of action remain poorly understood. Komatsu et al. show that Rolipram, a specific inhibitor of PDE4, reduces inflammation in a model of middle ear infection by upregulating the deubiquitinase CYLD.
Collapse
Affiliation(s)
- Kensei Komatsu
- Center for Inflammation, Immunity & Infection and Department of Biology, Georgia State University, 100 Piedmont Avenue, Atlanta, Georgia 30303, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Li JD, Hermansson A, Ryan AF, Bakaletz LO, Brown SD, Cheeseman MT, Juhn SK, Jung TTK, Lim DJ, Lim JH, Lin J, Moon SK, Post JC. Panel 4: Recent advances in otitis media in molecular biology, biochemistry, genetics, and animal models. Otolaryngol Head Neck Surg 2013; 148:E52-63. [PMID: 23536532 DOI: 10.1177/0194599813479772] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Otitis media (OM) is the most common childhood bacterial infection and also the leading cause of conductive hearing loss in children. Currently, there is an urgent need for developing novel therapeutic agents for treating OM based on full understanding of molecular pathogenesis in the areas of molecular biology, biochemistry, genetics, and animal model studies in OM. OBJECTIVE To provide a state-of-the-art review concerning recent advances in OM in the areas of molecular biology, biochemistry, genetics, and animal model studies and to discuss the future directions of OM studies in these areas. DATA SOURCES AND REVIEW METHODS A structured search of the current literature (since June 2007). The authors searched PubMed for published literature in the areas of molecular biology, biochemistry, genetics, and animal model studies in OM. RESULTS Over the past 4 years, significant progress has been made in the areas of molecular biology, biochemistry, genetics, and animal model studies in OM. These studies brought new insights into our understanding of the molecular and biochemical mechanisms underlying the molecular pathogenesis of OM and helped identify novel therapeutic targets for OM. CONCLUSIONS AND IMPLICATIONS FOR PRACTICE Our understanding of the molecular pathogenesis of OM has been significantly advanced, particularly in the areas of inflammation, innate immunity, mucus overproduction, mucosal hyperplasia, middle ear and inner ear interaction, genetics, genome sequencing, and animal model studies. Although these studies are still in their experimental stages, they help identify new potential therapeutic targets. Future preclinical and clinical studies will help to translate these exciting experimental research findings into clinical applications.
Collapse
Affiliation(s)
- Jian-Dong Li
- Center for Inflammation, Immunity & Infection, and Department of Biology, Georgia State University, Atlanta, Georgia 30303, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|