Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 134(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38051587

ABSTRACT

Hidradenitis suppurativa (HS) is a chronic inflammatory disease characterized by abscesses, nodules, dissecting/draining tunnels, and extensive fibrosis. Here, we integrate single-cell RNA sequencing, spatial transcriptomics, and immunostaining to provide an unprecedented view of the pathogenesis of chronic HS, characterizing the main cellular players and defining their interactions. We found a striking layering of the chronic HS infiltrate and identified the contribution of 2 fibroblast subtypes (SFRP4+ and CXCL13+) in orchestrating this compartmentalized immune response. We further demonstrated the central role of the Hippo pathway in promoting extensive fibrosis in HS and provided preclinical evidence that the profibrotic fibroblast response in HS can be modulated through inhibition of this pathway. These data provide insights into key aspects of HS pathogenesis with broad therapeutic implications.


Subject(s)
Hidradenitis Suppurativa , Humans , Hidradenitis Suppurativa/genetics , Hippo Signaling Pathway , Fibrosis
2.
Eur J Pharmacol ; 945: 175533, 2023 Apr 15.
Article in English | MEDLINE | ID: mdl-36690055

ABSTRACT

BACKGROUND: Histamine has been postulated to play a role in atopic dermatitis via histamine receptor 4, mediating pruritic and inflammatory effects. The H4R antagonist adriforant (PF-3893787 or ZPL389) indicated clinical efficacy in a Ph2a study in atopic dermatitis. Preclinical investigations of adriforant had been scarce as experiments in transfectants with H4R from several species suggested partial agonism, not seen in human cells. OBJECTIVE: During the Ph2b trial in AD, we performed experiments to understand the pharmacology of adriforant in primary murine cells and in vivo models. We assessed its effects on ERK phosphorylation and transcriptional changes in bone marrow-derived mast cells, histamine-dependent Ca2+ flux in neurons and histamine-induced itch response. In addition, its impact on MC903-induced skin inflammation was evaluated. RESULTS: We show that, contrary to transfectants, adriforant is a competitive antagonist of the murine histamine receptor 4, antagonizes histamine-induced ERK phosphorylation, normalizes histamine-induced transcriptional changes in mast cells and reduces histamine-dependent Ca2+ flux in neurons. Administration to mice reduces acute histamine-induced itch response. In addition, adriforant ameliorates inflammation in the mouse MC903 model. CONCLUSIONS: Our results suggest that functional inhibition of histamine receptor 4 by adriforant reduces itch and inflammation in vivo. The effects observed in mice, however, did not translate to clinical efficacy in patients as the Ph2b clinical trial with adriforant did not meet pre-specified efficacy endpoints. Given the complex pathogenesis of AD, antagonism of histamine receptor 4 alone appears insufficient to reduce disease severity in AD patients, despite the effects seen in mouse models.


Subject(s)
Dermatitis, Atopic , Humans , Mice , Animals , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/chemically induced , Histamine/pharmacology , Pruritus/chemically induced , Pruritus/drug therapy , Receptors, Histamine , Inflammation/drug therapy , Skin
3.
Lancet Microbe ; 3(4): e274-e283, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35165669

ABSTRACT

BACKGROUND: Vaccination is an efficient strategy to control the COVID-19 pandemic. In north Cyprus, vaccine distribution started with CoronaVac followed by BNT162b2, and ChAdOx1 vaccines. An option to obtain a third booster dose with BNT162b2 or CoronaVac was later offered to people fully inoculated with CoronaVac. There are few simultaneous and comparative real-world antibody data for these three vaccines as well as boosters after CoronaVac vaccination. Our study was aimed at evaluating antibody responses after these vaccination schemes. METHODS: We did a prospective, longitudinal population-based study to measure SARS-CoV-2 anti-spike receptor binding domain (RBD) IgG concentrations, assessed by assaying blood samples collected, in participants in north Cyprus who had received the BNT162b2, ChAdOx1, or CoronaVac vaccine at 1 month and 3 months after the second dose. Participants were recruited when they voluntarily came to the laboratory for testing after vaccination, solicited from health-care access points, or from the general population. We also evaluated antibody responses 1 month after a booster dose of BNT162b2 or CoronaVac after primary CoronaVac regimen. Demographics, baseline characteristics, vaccination reactions, and percentage of antibody responders were collected by phone interviews or directly from the laboratory summarised by vaccine and age group. Antibody levels were compared between groups over time by parametric and non-parametric methods. FINDINGS: Recruitment, follow-up, and data collection was done between March 1 and Sept 30, 2021. BNT162b2 induced the highest seropositivity and anti-spike RBD IgG antibody titres, followed by ChAdOx1, and then by CoronaVac. In addition, the rate of decline of antibodies was fastest with CoronaVac, followed by ChAdOx1, and then by BNT162b2. For the older age group, the rate of seropositivity at 3 months after the second dose was 100% for BNT162b2, 90% for ChAdOx1, and 60% for CoronaVac. In the multivariate repeated measures model, lower antibody titres were also significantly associated with male sex, older age, and time since vaccination. Boosting a two-dose CoronaVac regimen at 6 months with a single BNT162b2 dose led to significantly increased titres of IgG compared with boosting with CoronaVac; for the 60 years and older age group, the geometric mean fold rise in antibody titre after the booster relative to 1 month post-baseline was 7·9 (95% CI 5·8-10·8) in the BNT162b2 boost group versus 2·8 (1·6-5·0) in the CoronaVac group. INTERPRETATION: These longitudinal data can help shape vaccination strategies. Given the low antibody titres and fast decline in the CoronaVac group in individuals 60 years or older, more potent vaccine options could be considered as the primary vaccination or booster dose in these high-risk populations to sustain antibody responses for longer. FUNDING: Crowdfunded in north Cyprus.


Subject(s)
COVID-19 , SARS-CoV-2 , Aged , Antibodies, Viral , Antibody Formation , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , ChAdOx1 nCoV-19 , Humans , Immunoglobulin G , Male , Pandemics , Prospective Studies
4.
J Allergy Clin Immunol ; 147(6): 2386-2393.e4, 2021 06.
Article in English | MEDLINE | ID: mdl-33675820

ABSTRACT

BACKGROUND: The molecular control of inflammation and epidermal thickening in skin lesions of patients with atopic dermatitis (AD) is not known. Sequestosome 1/p62 is a multifunctional adapter protein implicated in the control of key regulators of cellular homeostasis, such as proinflammatory and mechanistic target of rapamycin signaling. OBJECTIVE: We sought to determine whether p62 plays a role in the cutaneous and systemic manifestations of an AD-like mouse model. METHODS: AD-like skin lesions were induced by deletion of JunB/AP-1, specifically in epidermal keratinocytes (JunBΔep). The contribution of p62 to pathological changes was determined by inactivation of p62 in JunBΔepp62-/- double knockout mice. RESULTS: Expression of p62 was elevated in skin lesions of JunBΔep mice, resembling upregulation of p62 in AD and psoriasis. When p62 was inactivated, JunBΔep-associated defects in the differentiation of keratinocytes, epidermal thickening, skin infiltration by mast cells and neutrophils, and the development of macroscopic skin lesions were significantly reduced. p62 inactivation had little effect on circulating cytokines, but decreased serum IgE. Signaling through mechanistic target of rapamycin and natural factor kappa B was increased in JunBΔep but not in JunBΔepp62-/- double knockout skin, indicating an important role of p62 in enhancing these signaling pathways in the skin during AD-like inflammation. CONCLUSIONS: Our results provide the first in vivo evidence for a proinflammatory role of p62 in skin and suggest that p62-dependent signaling pathways may be promising therapeutic targets to ameliorate the skin manifestations of AD and possibly psoriasis.


Subject(s)
Dermatitis, Atopic/etiology , Dermatitis, Atopic/metabolism , Disease Susceptibility , Sequestosome-1 Protein/metabolism , Animals , Biomarkers , Chronic Disease , Dermatitis, Atopic/pathology , Disease Models, Animal , Mice , Mice, Knockout , Phenotype , Sequestosome-1 Protein/genetics , Signal Transduction , Skin/immunology , Skin/metabolism , Skin/pathology
5.
J Allergy Clin Immunol ; 148(3): 799-812.e10, 2021 09.
Article in English | MEDLINE | ID: mdl-33662369

ABSTRACT

BACKGROUND: The type 2 cytokines IL-4 and IL-13 promote not only atopic dermatitis (AD) but also the resolution of inflammation. How type 2 cytokines participate in the resolution of AD is poorly known. OBJECTIVE: Our aim was to determine the mechanisms and cell types governing skin inflammation, barrier dysfunction, and resolution of inflammation in a model of AD. METHODS: Mice that exhibit expression of IL-4, IL-13, and MCPT8 or that could be depleted of basophils or eosinophils, be deficient in IL-4 or MHC class II molecules, or have basophils lacking macrophage colony-stimulating factor (M-CSF) were treated with calcipotriol (MC903) as an acute model of AD. Kinetics of the disease; keratinocyte differentiation; and leukocyte accumulation, phenotype, function, and cytokine production were measured by transepidermal water loss, histopathology, molecular biology, or unbiased analysis of spectral flow cytometry. RESULTS: In this model of AD, basophils were activated systemically and were the initial and main source of IL-4 in the skin. Basophils and IL-4 promoted epidermal hyperplasia and skin barrier dysfunction by acting on keratinocyte differentiation during inflammation. Basophils, IL-4, and basophil-derived M-CSF inhibited the accumulation of proinflammatory cells in the skin while promoting the expansion and function of proresolution M2-like macrophages and the expression of probarrier genes. Basophils kept their proresolution properties during AD resolution. CONCLUSION: Basophils can display both beneficial and detrimental type 2 functions simultaneously during atopic inflammation.


Subject(s)
Basophils/immunology , Dermatitis, Atopic/immunology , Skin/immunology , Animals , Calcitriol/analogs & derivatives , Cell Differentiation , Cytokines/genetics , Cytokines/immunology , Dermatitis, Atopic/chemically induced , Dermatitis, Atopic/genetics , Dermatitis, Atopic/pathology , Diphtheria Toxin , Edema/chemically induced , Edema/immunology , Eosinophils/immunology , Female , Gene Expression , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Hyperplasia/immunology , Keratinocytes/cytology , Male , Mice, Inbred C57BL , Mice, Transgenic , Skin/pathology
6.
Cell Rep ; 29(4): 844-859.e3, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31644908

ABSTRACT

Atopic dermatitis (AD) is a multi-factorial skin disease with a complex inflammatory signature including type 2 and type 17 activation. Although colonization by S. aureus is common in AD, the mechanisms rendering an organism prone to dysbiosis, and the role of IL-17A in the control of S. aureus-induced skin inflammation, are not well understood. Here, we show several pathological aspects of AD, including type 2/type 17 immune responses, elevated IgE, barrier dysfunction, pruritus, and importantly, spontaneous S. aureus colonization in JunBΔep mice, with a large transcriptomic overlap with AD. Additionally, using Rag1-/- mice, we demonstrate that adaptive immune cells are necessary for protection against S. aureus colonization. Prophylactic antibiotics, but not antibiotics after established dysbiosis, reduce IL-17A expression and skin inflammation, examined using Il17a-eGFP reporter mice. Mechanistically, keratinocytes lacking JunB exhibit higher MyD88 levels in vitro and in vivo, previously shown to regulate S. aureus colonization. In conclusion, our data identify JunB as an upstream regulator of microbiota-immune cell interactions and characterize the IL-17A response upon spontaneous dysbiosis.


Subject(s)
Dermatitis, Atopic/immunology , Epidermis/microbiology , Microbiota , Staphylococcal Infections/immunology , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Animals , Dermatitis, Atopic/complications , Dermatitis, Atopic/microbiology , Epidermis/immunology , Female , Homeodomain Proteins/genetics , Immunoglobulin E/immunology , Interleukin-17/genetics , Interleukin-17/metabolism , Male , Mice , Mice, Inbred C57BL , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity
7.
Methods Mol Biol ; 1914: 343-348, 2019.
Article in English | MEDLINE | ID: mdl-30729475

ABSTRACT

Melanomas are aggressive cancers of the skin with high metastatic capacity. Mouse models are necessary to delineate the mechanisms of cancer metastasis and xenograft models can also allow examining the role of the host using different genetically-modified mouse models. In this chapter, I provide a detailed protocol for the preparation and inoculation of tumor cells intra-cardially and intra-tibially to achieve bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Melanoma, Experimental/pathology , Skin Neoplasms/pathology , Animals , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/pathology , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Line, Tumor/transplantation , Humans , Luciferases/chemistry , Luminescent Measurements/instrumentation , Luminescent Measurements/methods , Mice , Mice, Inbred C57BL , Optical Imaging/instrumentation , Optical Imaging/methods , Tibia/diagnostic imaging , Tibia/pathology
8.
Mol Cell Biol ; 38(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30181393

ABSTRACT

Inflammatory responses require mobilization of innate immune cells from the bone marrow. The functionality of this process depends on the state of the bone marrow microenvironment. We therefore hypothesized that molecular changes in osteoblasts, which are essential stromal cells of the bone marrow microenvironment, influence the inflammatory response. Here, we show that osteoblast-specific expression of the AP-1 transcription factor Fra-2 (Fra-2Ob-tet) induced a systemic inflammatory state with infiltration of neutrophils and proinflammatory macrophages into the spleen and liver as well as increased levels of proinflammatory cytokines, such as interleukin-1ß (IL-1ß), IL-6, and granulocyte-macrophage colony-stimulating factor (GM-CSF). By in vivo inhibition of osteopontin (OPN) in Fra-2Ob-tet mice, we demonstrated that this process was dependent on OPN expression, which mediates alterations of the bone marrow niche. OPN expression was transcriptionally enhanced by Fra-2 and stimulated mesenchymal stem cell (MSC) expansion. Furthermore, in a murine lung injury model, Fra-2Ob-tet mice showed increased inflammatory responses and more severe disease features via an enhanced and sustained inflammatory response to lipopolysaccharide (LPS). Our findings demonstrate for the first time that molecular changes in osteoblasts influence the susceptibility to inflammation by altering evasion of innate immune cells from the bone marrow space.


Subject(s)
Fos-Related Antigen-2/metabolism , Inflammation/metabolism , Lung Injury/metabolism , Osteoblasts/metabolism , Osteopontin/metabolism , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , Cells, Cultured , Cytokines/metabolism , Immunity, Innate/immunology , Inflammation/immunology , Lipopolysaccharides/immunology , Lung Injury/immunology , Mice , Neutrophils/immunology , Neutrophils/metabolism , Osteoblasts/immunology , Transcription Factor AP-1/metabolism
9.
Nat Commun ; 9(1): 1466, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29654229

ABSTRACT

To accurately recapitulate the heterogeneity of human diseases, animal models require to recreate multiple complex genetic alterations. Here, we combine the RCAS-TVA system with the CRISPR-Cas9 genome editing tools for precise modeling of human tumors. We show that somatic deletion in neural stem cells of a variety of known tumor suppressor genes (Trp53, Cdkn2a, and Pten) leads to high-grade glioma formation. Moreover, by simultaneous delivery of pairs of guide RNAs we generate different gene fusions with oncogenic potential, either by chromosomal deletion (Bcan-Ntrk1) or by chromosomal translocation (Myb-Qk). Lastly, using homology-directed-repair, we also produce tumors carrying the homologous mutation to human BRAF V600E, frequently identified in a variety of tumors, including different types of gliomas. In summary, we have developed an extremely versatile mouse model for in vivo somatic genome editing, that will elicit the generation of more accurate cancer models particularly appropriate for pre-clinical testing.


Subject(s)
Brain Neoplasms/genetics , CRISPR-Cas Systems , Gene Editing , RNA, Guide, Kinetoplastida/genetics , Animals , Antigens, Neoplasm/genetics , Benzamides/pharmacology , Brain Neoplasms/drug therapy , Brevican/genetics , DNA Repair , False Positive Reactions , Gene Frequency , Gene Transfer Techniques , Glioma/metabolism , Humans , In Situ Hybridization, Fluorescence , Indazoles/pharmacology , Mice , Mice, SCID , Mice, Transgenic , Mutation , NIH 3T3 Cells , Receptor, trkA/genetics
10.
FEBS J ; 284(4): 505-516, 2017 02.
Article in English | MEDLINE | ID: mdl-27650997

ABSTRACT

Chronic systemic inflammation (CSI) has recently been identified as a major contributor to common diseases ranging from cancer to metabolic disorders and neurologic alterations. In the last decade, we and others have generated genetically engineered mouse models for inflammatory diseases, which enable studying the molecular mechanisms of CSI. Recently, organ cross-talk induced by CSI under homeostatic and pathological conditions has begun to be appreciated. In this review, we will revisit whole organism physiology in relation to CSI originating from epithelial tissues, such as the skin and gut. Furthermore, we will discuss the current knowledge regarding the mechanisms, the specific immune cells and molecules responsible for inducing the most common comorbidities, such as cardiovascular, metabolic, and neurological complications, as well as bone loss, in heterogeneous diseases like psoriasis, atopic dermatitis, and inflammatory bowel disease. As it would be impossible to discuss all comorbidities of these diseases as well as all epithelial tissues, we present an overview with a special emphasis on our recent findings linking skin inflammation to bone loss.


Subject(s)
Atherosclerosis/immunology , Bone Resorption/immunology , Dermatitis, Atopic/immunology , Epithelium/immunology , Inflammatory Bowel Diseases/immunology , Psoriasis/immunology , Animals , Atherosclerosis/complications , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Resorption/complications , Bone Resorption/genetics , Bone Resorption/pathology , Chronic Disease , Dermatitis, Atopic/complications , Dermatitis, Atopic/genetics , Dermatitis, Atopic/pathology , Disease Models, Animal , Epithelium/pathology , Gene Expression , Humans , Inflammation , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukins/genetics , Interleukins/immunology , Mice , Mice, Transgenic , Psoriasis/complications , Psoriasis/genetics , Psoriasis/pathology
11.
Clin Exp Rheumatol ; 34(4 Suppl 98): 17-20, 2016.
Article in English | MEDLINE | ID: mdl-27586798

ABSTRACT

Inflammation is a physiological reaction to tissue injury, pathogen invasion and a natural response to various stress stimuli. Innate and adaptive immune cells are activated and recruited to the site of inflammation to suppress or promote inflammation. The recruitment and activation of immune cells is modulated by cytokines and chemokines, which are regulated by transcription factors, such as AP-1 (Fos/Jun), NF-kB, NFATs and STATs. Moreover, it is now appreciated that chronic inflammation can lead to systemic effects affecting the whole organism by mechanisms which are not well understood.Here we review our recent data obtained from the analyses of psoriasis patient samples as well as from AP-1 (Fos/Jun)-dependent, genetically engineered mouse models. The deletion of two AP-1 factors JunB and c-Jun in an inducible manner in adult mice, specifically in Keratin-5 expressing tissues, leads to a psoriasis-like disease. Importantly, the epidermal proteome of the mutant mice is comparable to psoriasis patient samples. Our analyses revealed that the activation of S100A8/A9-dependent C3 complement as well as a miR-21-dependent TIMP-3/TACE pathway leading to TNF-α shedding, are causally involved in disease development.Epidermal deletion of only JunB in mice leads to chronic skin inflammation with increased levels of pro-inflammatory cytokines and multi-organ involvement. Our recent findings show that chronic skin inflammation induces bone loss through systemic elevated IL-17A signalling. This novel mechanism involves inhibition of osteoblast-mediated bone formation by reduced Wnt signalling with no effect on RANKL-dependent osteoclastic bone resorption. These data have important translational implications; blocking of IL-17A signalling, which is already approved for the treatment of psoriasis, should also be considered to prevent the adverse skeletal consequences of chronic inflammatory diseases.


Subject(s)
Bone Resorption/metabolism , Bone and Bones/metabolism , Inflammation Mediators/metabolism , Interleukin-17/metabolism , Psoriasis/metabolism , Skin/metabolism , Wnt Signaling Pathway , Animals , Bone Resorption/genetics , Bone Resorption/immunology , Bone Resorption/pathology , Bone and Bones/immunology , Bone and Bones/pathology , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Inflammation Mediators/immunology , Interleukin-17/immunology , Mice, Transgenic , Phenotype , Psoriasis/genetics , Psoriasis/immunology , Psoriasis/pathology , Skin/immunology , Skin/pathology
12.
Sci Transl Med ; 8(330): 330ra37, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-27089206

ABSTRACT

Inflammation has important roles in tissue regeneration, autoimmunity, and cancer. Different inflammatory stimuli can lead to bone loss by mechanisms that are not well understood. We show that skin inflammation induces bone loss in mice and humans. In psoriasis, one of the prototypic IL-17A-mediated inflammatory human skin diseases, low bone formation and bone loss correlated with increased serum IL-17A levels. Similarly, in two mouse models with chronic IL-17A-mediated skin inflammation,K14-IL17A(ind)andJunB(Δep), strong inhibition of bone formation was observed, different from classical inflammatory bone loss where osteoclast activation leads to bone degradation. We show that under inflammatory conditions, skin-resident cells such as keratinocytes, γδ T cells, and innate lymphoid cells were able to express IL-17A, which acted systemically to inhibit osteoblast and osteocyte function by a mechanism involving Wnt signaling. IL-17A led to decreased Wnt signaling in vitro, and importantly, pharmacological blockade of IL-17A rescued Wnt target gene expression and bone formation in vivo. These data provide a mechanism where IL-17A affects bone formation by regulating Wnt signaling in osteoblasts and osteocytes. This study suggests that using IL-17A blocking agents in psoriasis could be beneficial against bone loss in these patients.


Subject(s)
Bone Resorption/pathology , Inflammation/pathology , Interleukin-17/metabolism , Osteoblasts/metabolism , Osteoblasts/pathology , Skin/pathology , Wnt Signaling Pathway , Animals , Bone Resorption/genetics , Cell Lineage , Chronic Disease , Epithelium/pathology , Female , Gene Expression Regulation , Humans , Inflammation/genetics , Male , Mice, Inbred C57BL , Middle Aged , Models, Biological , Osteocytes/metabolism , Osteocytes/pathology , Osteogenesis , Psoriasis
13.
Clin Exp Rheumatol ; 33(4 Suppl 92): S44-9, 2015.
Article in English | MEDLINE | ID: mdl-26458100

ABSTRACT

Skin inflammation is a physiological reaction to tissue injury, pathogen invasion and irritants. During this process, innate and/or adaptive immune cells are activated and recruited to the site of inflammation to either promote or suppress inflammation. The sequential recruitment and activation of immune cells is modulated by a combination of cytokines and chemokines, which are regulated by transcription factors, such as AP-1 (Fos/Jun), NF-κB, NFATs, and STATs. Here we review the present evidence and the underlying mechanisms of how Jun/AP-1 proteins control skin inflammation. Genetically engineered mouse models (GEMMs) in which AP-1 proteins are deleted in the epidermis have revealed that these proteins control cytokine expression at multiple levels. Constitutive epidermal deletion of JunB in mice leads to a multi-organ disease characterised by increased levels of pro-inflammatory cytokines. These JunB-deficient mutant mice display several phenotypes from skin inflammation to a G-CSF-dependent myeloproliferative disease, as well as kidney atrophy and bone loss, reminiscent of psoriasis and systemic lupus erythematosus. Importantly, epidermal deletion of both JunB and c-Jun in an inducible manner in adult mice leads to a psoriasis-like disease, in which the epidermal proteome expression profile is comparable to the one from psoriasis patient samples. In this GEMM and in psoriasis patient-derived material, S100A8/A9-dependent C3/CFB complement activation, as well as a miR-21-dependent TIMP-3/TACE pathway leading to TNF-α shedding, plays causal roles in disease development. The newly identified therapeutic targets from GEMMs together with investigations in human patient samples open up new avenues for therapeutic interventions for psoriasis and related inflammatory skin diseases.


Subject(s)
Dermatitis/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Skin/metabolism , Transcription Factor AP-1/metabolism , Animals , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/pathology , Gene Expression Regulation , Genotype , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Phenotype , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-jun/genetics , Skin/immunology , Skin/pathology , Transcription Factor AP-1/genetics
14.
Bonekey Rep ; 4: 670, 2015.
Article in English | MEDLINE | ID: mdl-25987985

ABSTRACT

Osteosarcoma is the most common form of primary bone tumors with high prevalence in children. Survival rates of osteosarcoma are low, especially in the case of metastases. Mouse models of this disease have been very valuable in investigation of mechanisms of tumorigenesis, metastasis, as well as testing possible therapeutic options. In this chapter, we summarize currently available mouse models for osteosarcoma and provide detailed methodology for the isolation of cell lines from genetically engineered mouse models (GEMMs), gene modification and tumor cell injection methods, as well as imaging techniques.

15.
Methods Mol Biol ; 1267: 297-305, 2015.
Article in English | MEDLINE | ID: mdl-25636475

ABSTRACT

Osteosarcoma (OS) is the most common primary tumor of bone with a high incidence in children. Treatment options for OS are limited, and once metastasized, the prognosis is very poor. Genetically engineered mouse models (GEMMs) are valuable tools to understand the mechanisms of tumorigenesis and to test possible therapies. In this chapter, we summarize the methods related to the isolation, characterization, and transplantation of OS cells obtained from GEMMs.


Subject(s)
Disease Models, Animal , Osteosarcoma/pathology , Alkaline Phosphatase/metabolism , Animals , Anthraquinones/metabolism , Bone Neoplasms/pathology , Carcinogenesis , Cell Line, Tumor , Cell Separation , Cell Transformation, Neoplastic , Mice , Staining and Labeling
16.
J Bone Miner Res ; 30(1): 106-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25042340

ABSTRACT

Thrombospondin-1 (TSP1), an endogenous antiangiogenic, is a widely expressed secreted ligand with roles in migration, adhesion, and proliferation and is a target for new therapeutics. While TSP1 is present in the bone matrix and several TSP1 receptors play roles in bone biology, the role of TSP1 in bone remodeling has not been fully elucidated. Bone turnover is characterized by coordinated activity of bone-forming osteoblasts (OB) and bone-resorbing osteoclasts (OC). TSP1-/- mice had increased bone mass and increased cortical bone size and thickness compared to wild type (WT). However, despite increased size, TSP1-/- femurs showed less resistance to bending than expected, indicative of diminished bone quality and a bone material defect. Additionally, we found that TSP1 deficiency resulted in decreased OC activity in vivo and reduced OC differentiation. TSP1 was critical during early osteoclastogenesis, and TSP1 deficiency resulted in a substantial overexpression of inducible nitric oxide synthase (iNOS). Importantly, administration of a NOS inhibitor rescued the OC function defects of TSP1-/- mice in vivo. To investigate the role of bone-derived TSP1 in osteoclastogenesis, we found that WT pre-OCs had defective iNOS expression when cultured on TSP1-/- bone compared to WT bone, suggesting that TSP1 in bone plays a critical role in iNOS signaling during OC development. These data implicate a new role for TSP1 in bone homeostasis with roles in maintaining bone matrix integrity and regulating OC formation. It will be critical to monitor bone health of patients administered TSP1-pathway directed therapeutics in clinical use and under development.


Subject(s)
Bone Matrix/metabolism , Nitric Oxide/metabolism , Osteoclasts/metabolism , Signal Transduction/physiology , Thrombospondin 1/metabolism , Animals , Bone Matrix/cytology , Cell Differentiation/physiology , Gene Expression Regulation, Enzymologic/genetics , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Osteoclasts/cytology , Thrombospondin 1/genetics
17.
Cell Cycle ; 13(12): 1980-6, 2014.
Article in English | MEDLINE | ID: mdl-24762558

ABSTRACT

Stable Fos expression in the osteoblast lineage results in the development of osteosarcomas (OS) in mice, yet the underlying mechanisms are poorly understood. Using a genetic system in which Fos expression can be induced in osteoblasts in a doxycycline-dependent manner and through subsequent RNA sequencing and gene set enrichment analysis, we were able to identify novel transcriptional targets of Fos in osteoblasts. These included a distinct activation of cellular response toward replication stress (RS), exemplified by a Fos-dependent induction of the RS-suppressing Chk1 kinase. Importantly, Fos expression protects osteoblasts from RS and DNA damage likely through upregulation of Chk1 and facilitates transformation by Ras/E1A oncogenes. These data reveal a novel function of Fos in safeguarding genome stability during replication, which is particularly relevant in conditions of oncogene-induced S-phase entry.


Subject(s)
DNA Replication/physiology , Oncogene Proteins v-fos/metabolism , Osteoblasts/metabolism , Protein Kinases/metabolism , Animals , Cells, Cultured , Checkpoint Kinase 1 , DNA Damage/physiology , Gene Knockout Techniques , Mice , Oncogene Proteins v-fos/genetics , Tumor Suppressor Protein p53/metabolism
18.
J Clin Invest ; 122(10): 3579-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22996695

ABSTRACT

The adenosine diphosphate (ADP) receptor P2RY12 (purinergic receptor P2Y, G protein coupled, 12) plays a critical role in platelet aggregation, and P2RY12 inhibitors are used clinically to prevent cardiac and cerebral thrombotic events. Extracellular ADP has also been shown to increase osteoclast (OC) activity, but the role of P2RY12 in OC biology is unknown. Here, we examined the role of mouse P2RY12 in OC function. Mice lacking P2ry12 had decreased OC activity and were partially protected from age-associated bone loss. P2ry12-/- OCs exhibited intact differentiation markers, but diminished resorptive function. Extracellular ADP enhanced OC adhesion and resorptive activity of WT, but not P2ry12-/-, OCs. In platelets, ADP stimulation of P2RY12 resulted in GTPase Ras-related protein (RAP1) activation and subsequent αIIbß3 integrin activation. Likewise, we found that ADP stimulation induced RAP1 activation in WT and integrin ß3 gene knockout (Itgb3-/-) OCs, but its effects were substantially blunted in P2ry12-/- OCs. In vivo, P2ry12-/- mice were partially protected from pathologic bone loss associated with serum transfer arthritis, tumor growth in bone, and ovariectomy-induced osteoporosis: all conditions associated with increased extracellular ADP. Finally, mice treated with the clinical inhibitor of P2RY12, clopidogrel, were protected from pathologic osteolysis. These results demonstrate that P2RY12 is the primary ADP receptor in OCs and suggest that P2RY12 inhibition is a potential therapeutic target for pathologic bone loss.


Subject(s)
Adenosine Diphosphate/physiology , Bone Remodeling/physiology , Osteoclasts/physiology , Osteoporosis/physiopathology , Receptors, Purinergic P2Y12/physiology , Animals , Arthritis, Experimental/complications , Bone Neoplasms/complications , Bone Neoplasms/secondary , Bone Remodeling/drug effects , Bone Resorption/physiopathology , Carcinoma/complications , Carcinoma/secondary , Cell Adhesion/drug effects , Cells, Cultured/drug effects , Clopidogrel , Enzyme Activation/drug effects , Female , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoporosis/etiology , Osteoporosis/prevention & control , Ovariectomy , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Purinergic P2Y Receptor Antagonists/pharmacology , Purinergic P2Y Receptor Antagonists/therapeutic use , Receptors, Purinergic P2Y12/deficiency , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y12/genetics , Specific Pathogen-Free Organisms , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology , Ticlopidine/therapeutic use , rap1 GTP-Binding Proteins/drug effects
19.
FASEB J ; 24(4): 1117-27, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19933310

ABSTRACT

The purpose of this work was to determine platelet and myeloid cell-specific requirements for beta3-containing integrins in hemostasis, bone resorption, and tumor growth. LoxP-flanked mice were generated to study the conditional deletion of beta3-integrin in platelets [knockout in platelets (KOP)] and myeloid cells [knockout in myeloid (KOM)]. Using the beta3KOP and beta3KOM strains of mice, we studied the role of beta3-integrin in hemostasis, bone resorption, and subcutaneous tumor growth. Tissue-specific deletion of platelet beta3-integrins in beta3KOP mice did not affect bone mass but resulted in a severe bleeding phenotype. No growth difference of tumor xenografts or in neoangiogenesis were found in beta3KOP mice, in contrast to the defects observed in germline beta3(-/-) mice. Conditional deletion of myeloid beta3-integrins in beta3KOM mice resulted in osteopetrosis but had no effect on hemostasis or mortality. Tumor growth in beta3KOM mice was increased and accompanied by decreased macrophage infiltration, without increase in blood vessel number. Platelet beta3-integrin deficiency was sufficient to disrupt hemostasis but had no effect on bone mass or tumor growth. Myeloid-specific beta3-integrin deletion was sufficient to perturb bone mass and enhance tumor growth due to reduced macrophage infiltration in the tumors. These results suggest that beta3-integrins have cell-specific roles in complex biological processes.-Morgan, E. A., Schneider, J. G., Baroni, T. E., Uluçkan, O., Heller, E., Hurchla, M. A., Deng, H., Floyd, D., Berdy, A., Prior, J. L., Piwnica-Worms, D., Teitelbaum, S. L., Ross, F. P., Weilbaecher, K. N. Dissection of platelet and myeloid cell defects by conditional targeting of the beta3-integrin subunit.


Subject(s)
Blood Platelets/metabolism , Bone Resorption/metabolism , Hemostasis , Integrin beta3/metabolism , Macrophages/metabolism , Melanoma/metabolism , Animals , Blood Platelets/pathology , Bone Resorption/genetics , Bone Resorption/pathology , Cell Line, Tumor , Hemorrhage/genetics , Hemorrhage/metabolism , Hemorrhage/pathology , Humans , Integrin beta3/genetics , Macrophages/pathology , Melanoma/genetics , Melanoma/pathology , Mice , Mice, Knockout , Neoplasm Transplantation , Organ Specificity/genetics , Transplantation, Heterologous
20.
Cancer Res ; 69(7): 3196-204, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276363

ABSTRACT

CD47, also called integrin-associated protein, plays a critical role in the innate immune response and is an atypical member of the immunoglobulin superfamily that interacts with and activates beta3 integrins. beta3 integrin(-/-) mice have defective platelet and osteoclast function and are protected from bone metastasis. The role of CD47 in skeletal homeostasis and bone metastasis has not been described. CD47(-/-) mice had increased bone mass and defective osteoclast function in vivo. Although the number of functional osteoclasts formed by differentiating CD47(-/-) bone marrow macrophages was decreased, high doses of RANKL rescued differentiation and function of CD47(-/-) osteoclasts ex vivo and rescued the osteoclast defect in CD47(-/-) mice. Inhibition of nitric oxide (NO) synthase, which is expressed at higher levels in CD47(-/-) osteoclasts, also rescued the osteoclast defect in CD47(-/-) cells. We then examined the consequences of this osteoclast defect in bone metastasis. In a model of tumor metastasis to bone, bone tumor burden was decreased in the CD47(-/-) mice compared with wild-type (WT) controls, with no decrease in s.c. tumor growth in CD47(-/-) mice. There was decreased tumor-associated bone destruction in the CD47(-/-) mice compared with WT controls, consistent with a defect in osteoclast function that was not rescued by the presence of tumor. Our data show that CD47 regulates osteoclastogenesis, in part, via regulation of NO production, and its disruption leads to a decrease in tumor bone metastasis. CD47 is a novel therapeutic target to strengthen bone mass and diminish metastatic tumor growth in bone.


Subject(s)
Bone Neoplasms/secondary , CD47 Antigen/metabolism , Melanoma, Experimental/pathology , Animals , Bone Neoplasms/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Humans , Integrin beta3/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/secondary , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nitric Oxide Synthase/antagonists & inhibitors , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , RANK Ligand/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...