Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
Cytokine ; 173: 156447, 2024 01.
Article in English | MEDLINE | ID: mdl-38041875

ABSTRACT

Lung macrophages are the first line of defense against invading respiratory pathogens including SARS-CoV-2, yet activation of macrophage in the lungs can lead to hyperinflammatory immune response seen in severe COVID-19. Here we used human M1 and M2 polarized macrophages as a surrogate model of inflammatory and regulatory macrophages and explored whether immune complexes (IC) containing spike-specific IgG can trigger aberrant cytokine responses in macrophages in the lungs and associated lymph nodes. We show that IC of SARS-CoV-2 recombinant S protein coated with spike-specific monoclonal antibody induced production of Prostaglandin E2 (PGE2) in non-polarized (M0) and in M1 and M2-type polarized human macrophages only in the presence of D-dimer (DD), a fibrinogen degradation product, associated with coagulopathy in COVID-19. Importantly, an increase in PGE2 was also observed in macrophages activated with DD and IC of SARS-CoV-2 pseudovirions coated with plasma from hospitalized COVID-19 patients but not from healthy subjects. Overall, the levels of PGE2 in macrophages activated with DD and IC were as follows: M1≫M2>M0 and correlated with the levels of spike binding antibodies and not with neutralizing antibody titers. All three macrophage subsets produced similar levels of IL-6 following activation with DD+IC, however TNFα, IL-1ß, and IL-10 cytokines were produced by M2 macrophages only. Our study suggests that high titers of spike or virion containing IC in the presence of coagulation byproducts (DD) can promote inflammatory response in macrophages in the lungs and associated lymph nodes and contribute to severe COVID-19.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , Antigen-Antibody Complex/metabolism , Inflammation Mediators/metabolism , Dinoprostone/metabolism , COVID-19/metabolism , Macrophages/metabolism , Cytokines/metabolism
2.
PLoS Pathog ; 18(4): e1010468, 2022 04.
Article in English | MEDLINE | ID: mdl-35385545

ABSTRACT

An overreactive inflammatory response and coagulopathy are observed in patients with severe form of COVID-19. Since increased levels of D-dimer (DD) are associated with coagulopathy in COVID-19, we explored whether DD contributes to the aberrant cytokine responses. Here we show that treatment of healthy human monocytes with DD induced a dose dependent increase in production of pyrogenic mediator, Prostaglandin E2 (PGE2) and inflammatory cytokines, IL-6 and IL-8. The DD-induced PGE2 and inflammatory cytokines were enhanced significantly by co-treatment with immune complexes (IC) of SARS CoV-2 recombinant S protein or of pseudovirus containing SARS CoV-2 S protein (PVCoV-2) coated with spike-specific chimeric monoclonal antibody (MAb) containing mouse variable and human Fc regions. The production of PGE2 and cytokines in monocytes activated with DD and ICs was sensitive to the inhibitors of ß2 integrin and FcγRIIa, and to the inhibitors of calcium signaling, Mitogen-Activated Protein Kinase (MAPK) pathway, and tyrosine-protein kinase. Importantly, strong increase in PGE2 and in IL-6/IL-8/IL-1ß cytokines was observed in monocytes activated with DD in the presence of IC of PVCoV-2 coated with plasma from hospitalized COVID-19 patients but not from healthy donors. The IC of PVCoV-2 with convalescent plasma induced much lower levels of PGE2 and cytokines compared with plasma from hospitalized COVID-19 patients. PGE2 and IL-6/IL-8 cytokines produced in monocytes activated with plasma-containing IC, correlated well with the levels of spike binding antibodies and not with neutralizing antibody titers. Our study suggests that a combination of high levels of DD and high titers of spike-binding antibodies that can form IC with SARS CoV-2 viral particles might accelerate the inflammatory status of lung infiltrating monocytes leading to increased lung pathology in patients with severe form of COVID-19.


Subject(s)
COVID-19 , Monocytes , Animals , Antigen-Antibody Complex , COVID-19/therapy , Cytokines/metabolism , Dinoprostone/metabolism , Fibrin Fibrinogen Degradation Products , Humans , Immunization, Passive , Immunologic Factors/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Mice , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Serotherapy
3.
Mol Immunol ; 128: 139-149, 2020 12.
Article in English | MEDLINE | ID: mdl-33126082

ABSTRACT

Fever and inflammatory responses were observed in some subjects in early clinical trials of vaccines adjuvanted with muramyl dipeptide (MDP), a NOD2 agonist. Biosynthesis of Prostaglandin E2 (PGE2) that transmits febrile signals to the brain is controlled by an inducible enzyme, Cyclooxygenase 2 (COX-2). MDP alone was not sufficient to induce expression of COX-2 and PGE2 production in vitro. Conditioned medium prepared from Peripheral Blood Mononuclear Cells (PBMCs)-derived CD3-bead purified human T cells (TCM) dramatically increased COX2 gene transcription, COX-2 protein expression, and PGE2 production in MDP-treated monocytes. We explored epigenetic changes at the COX2 promoter using Chromatin Immunoprecipitation assay (ChIP). Increase in COX2 transcription correlated with increased recruitment of RNA polymerase II (Pol II) and p300 histone acetyl transferase (HAT) to the COX2 promoter in monocytes activated with MDP and TCM. The role of p300 HAT was confirmed by using C646, an inhibitor of p300, that reduced binding of acetylated H3 and H4 histones at the COX2 promoter, COX2 transcription, and PGE2 production in monocytes. Binding of p300, Nuclear Factor Kappa B (NF-κB), and Pol II to the COX2 promoter was also sensitive to inhibitors of Mitogen-Activated Protein Kinase (MAPK) pathway and to antibodies against Macrophage-1 (Mac-1) integrin in MDP/TCM-treated monocytes. Importantly, recombinant Glycoprotein Ib alfa (GPIbα), the recently identified factor in TCM, increased binding of NF-κB, p300, and of Pol II to the COX2 promoter and COX2 transcription in MDP-treated monocytes. Our findings suggest that a second signal through Mac-1 and MAPK is triggered by a T cell derived soluble GPIbα protein leading to the assembly of the transcription machinery at the COX2 promoter and production of PGE2 in human monocytes in response to MDP/NOD2 activation.


Subject(s)
Dinoprostone/metabolism , E1A-Associated p300 Protein/metabolism , Mitogen-Activated Protein Kinases/metabolism , Monocytes/metabolism , Signal Transduction/physiology , T-Lymphocytes/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Adjuvants, Immunologic/pharmacology , Cells, Cultured , Cyclooxygenase 2/metabolism , Fever/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Macrophage-1 Antigen/metabolism , Macrophages/drug effects , Macrophages/metabolism , Monocytes/drug effects , NF-kappa B/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Signal Transduction/drug effects , T-Lymphocytes/drug effects , Th1 Cells/drug effects , Th1 Cells/metabolism , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
4.
Sci Signal ; 12(602)2019 10 08.
Article in English | MEDLINE | ID: mdl-31594856

ABSTRACT

Vaccine adjuvants containing analogs of microbial products activate pattern recognition receptors (PRRs) on antigen-presenting cells, including monocytes and macrophages, which can cause prostaglandin E2 (PGE2) release and consequently undesired inflammatory responses and fever in vaccine recipients. Here, we studied the mechanism of PGE2 production by human monocytes activated with muramyl dipeptide (MDP) adjuvant, which activates cytosolic nucleotide-binding oligomerization domain 2 (NOD2). In rabbits, administration of MDP elicited an early increase in PGE2 followed by fever. In human monocytes, MDP alone did not induce PGE2 production. However, high amounts of PGE2 and the proinflammatory cytokines IL-1ß and IL-6 were secreted by monocytes activated with MDP in the presence of conditioned medium obtained from CD3 bead-isolated T cells (Tc CM) but not from those isolated without CD3 beads. Mass spectrometry and immunoblotting revealed that the costimulatory factor in Tc CM was glycoprotein Ib α (GPIbα). Antibody-mediated blockade of GPIbα or of its receptor, Mac-1 integrin, inhibited the secretion of PGE2, IL-1ß, and IL-6 in MDP + Tc CM-activated monocytes, whereas recombinant GPIbα protein increased PGE2 production by MDP-treated monocytes. In vivo, COX2 mRNA abundance was reduced in the liver and spleen of Mac-1 KO mice after administration of MDP compared with that of treated wild-type mice. Our findings suggest that the production of PGE2 and proinflammatory cytokines by MDP-activated monocytes is mediated by cooperation between two signaling pathways: one delivered by MDP through NOD2 and a second through activation of Mac-1 by T cell-derived GPIbα.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Dinoprostone/metabolism , Monocytes/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , T-Lymphocytes/metabolism , Adjuvants, Immunologic/pharmacology , Animals , Calcium/metabolism , Cells, Cultured , Culture Media, Conditioned/pharmacology , Female , HEK293 Cells , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophage-1 Antigen/genetics , Macrophage-1 Antigen/metabolism , Mice, Knockout , Monocytes/cytology , Monocytes/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Platelet Glycoprotein GPIb-IX Complex/genetics , Rabbits , Signal Transduction/drug effects , THP-1 Cells
5.
Article in English | MEDLINE | ID: mdl-28348035

ABSTRACT

Animal models have played a pivotal role in all stages of vaccine development. Their predictive value for vaccine effectiveness depends on the pathogen, the robustness of the animal challenge model, and the correlates of protection (if known). This article will cover key questions regarding bridging animal studies to efficacy trials in humans. Examples include human papillomavirus (HPV) vaccine in which animal protection after vaccination with heterologous prototype virus-like particles (VLPs) predicted successful efficacy trials in humans, and a recent approval of anthrax vaccine in accordance with the "Animal Rule." The establishment of animal models predictive of vaccine effectiveness in humans has been fraught with difficulties with low success rate to date. Challenges facing the use of animal models for vaccine development against Ebola and HIV will be discussed.


Subject(s)
Disease Models, Animal , Vaccines/therapeutic use , AIDS Vaccines/adverse effects , AIDS Vaccines/therapeutic use , Animals , Anthrax/immunology , Anthrax Vaccines/adverse effects , Anthrax Vaccines/therapeutic use , Drug Approval , Drug Evaluation, Preclinical , Ebola Vaccines/adverse effects , Ebola Vaccines/therapeutic use , Humans , Papillomaviridae/immunology , Papillomavirus Vaccines/adverse effects , Papillomavirus Vaccines/therapeutic use , Proof of Concept Study , Species Specificity
6.
Antiviral Res ; 144: 8-20, 2017 08.
Article in English | MEDLINE | ID: mdl-28495463

ABSTRACT

Bioluminescence imaging (BLI) was used to follow dissemination of recombinant vaccinia virus (VACV) expressing luciferase (IHD-J-Luc) in BALB/c nu/nu mice treated post-challenge with monoclonal antibodies (MAbs) against L1 and B5 VACV proteins in a model of Progressive Vaccinia (PV). Areas Under the flux Curve (AUC) were calculated for viral loads in multiple organs in individual mice. Following scarification with 105 pfu, IHD-J-Luc VACV undergoes fast replication at the injection site and disseminates rapidly to the inguinal lymph nodes followed by spleen, liver, and axillary lymph nodes within 2-3 days and before primary lesions are visible at the site of scarification. Extension of survival in nude mice treated with a combination of anti-B5 and anti-L1 MAbs 24 h post challenge correlated with a significant reduction in viral load at the site of scarification and delayed systemic dissemination. Nude mice reconstituted with 104 T cells prior to challenge with IHD-J-Luc, and treated with MAbs post-challenge, survived infection, cleared the virus from all organs and scarification site, and developed anti-VACV IgG and VACV-specific polyfunctional CD8+ T cells that co-expressed the degranulation marker CD107a, and IFNγ and TNFα cytokines. All T cell reconstituted mice survived intranasal re-challenge with IHD-J-Luc (104 pfu) two months after the primary infection. Thus, using BLI to monitor VACV replication in a PV model, we showed that anti-VACV MAbs administered post challenge extended survival of nude mice and protected T cell reconstituted nude mice from lethality by reducing replication at the site of scarification and systemic dissemination of VACV.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antiviral Agents/administration & dosage , Disease Models, Animal , Vaccinia virus/growth & development , Vaccinia/pathology , Vaccinia/therapy , Animal Structures/virology , Animals , Immunologic Factors/administration & dosage , Luminescent Measurements , Mice, Inbred BALB C , Mice, Nude , Survival Analysis , Treatment Outcome , Viral Load , Viral Proteins/immunology , Whole Body Imaging
7.
Radiat Environ Biophys ; 55(2): 185-94, 2016 May.
Article in English | MEDLINE | ID: mdl-26994995

ABSTRACT

The xCELLigence real-time cell impedance system uses a non-invasive and label-free method to create a cell index that is a composite measure of cell proliferation. The aim of this study was to evaluate xCELLigence against clonogenic assay (gold standard) for measuring radiobiological effects and radiation-induced bystander effects (RIBE). A radiobiological study was conducted by irradiating EMT6.5, 4T1.2 and NMUMG cell lines with different radiation doses, while a RIBE study was done using transfer of conditioned media (CM) harvested from donor to the same type of recipient cell (EMT6.5, 4T1.2, NMUMG, HACAT and SW48). CM was harvested using two protocols which differed in the dose chosen and the exposure to the recipient cells. Results showed that xCELLigence measured a radiobiological effect which correlated with the clonogenic assay. For the RIBE study, no statistically significant differences were observed between xCELLigence or clonogenic survival in control or recipient cells incubated with CM in protocol one. However, there was a significant increase in cell index slope using CM from EMT-6.5 cells irradiated at 7.5 Gy compared with the control group under the second protocol. No other evidence of RIBE was detected by either xCELLigence or clonogenic assay. In conclusion, xCELLigence methods can measure radiobiological effects and the results correlate with clonogenic assay. We observed a lack of RIBE in all tested cell lines with the clonogenic assay; however, we observed a RIBE effect in EMT6.5 cells under one particular protocol that showed RIBE is cell type dependent, is not universally observed and can be detected in different assays.


Subject(s)
Bystander Effect/radiation effects , Radiobiology/methods , Animals , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Culture Media, Conditioned , Dose-Response Relationship, Radiation , Humans , Mice , Reproducibility of Results , Time Factors
8.
J Virol ; 89(6): 3295-307, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25589648

ABSTRACT

UNLABELLED: Protection from lethality by postchallenge administration of brincidofovir (BCV, CMX001) was studied in normal and immune-deficient (nude, nu/nu) BALB/c mice infected with vaccinia virus (VACV). Whole-body bioluminescence imaging was used to record total fluxes in the nasal cavity, lungs, spleen, and liver and to enumerate pox lesions on tails of mice infected via the intranasal route with 10(5) PFU of recombinant IHD-J-Luc VACV expressing luciferase. Areas under the flux curve (AUCs) were calculated for individual mice to assess viral loads. A three-dose regimen of 20 mg/kg BCV administered every 48 h starting either on day 1 or day 2 postchallenge protected 100% of mice. Initiating BCV treatment earlier was more efficient in reducing viral loads and in providing protection from pox lesion development. All BCV-treated mice that survived challenge were also protected from rechallenge with IHD-J-Luc or WRvFire VACV without additional treatment. In immune-deficient mice, BCV protected animals from lethality and reduced viral loads while animals were on the drug. Viral recrudescence occurred within 4 to 9 days, and mice succumbed ∼10 to 20 days after treatment termination. Nude mice reconstituted with 10(5) T cells prior to challenge with 10(4) PFU of IHD-J-Luc and treated with BCV postchallenge survived the infection, cleared the virus from all organs, and survived rechallenge with 10(5) PFU of IHD-J-Luc VACV without additional BCV treatment. Together, these data suggest that BCV protects immunocompetent and partially T cell-reconstituted immune-deficient mice from lethality, reduces viral dissemination in organs, prevents pox lesion development, and permits generation of VACV-specific memory. IMPORTANCE: Mass vaccination is the primary element of the public health response to a smallpox outbreak. In addition to vaccination, however, antiviral drugs are required for individuals with uncertain exposure status to smallpox or for whom vaccination is contraindicated. Whole-body bioluminescence imaging was used to study the effect of brincidofovir (BCV) in normal and immune-deficient (nu/nu) mice infected with vaccinia virus, a model of smallpox. Postchallenge administration of 20 mg/kg BCV rescued normal and immune-deficient mice partially reconstituted with T cells from lethality and significantly reduced viral loads in organs. All BCV-treated mice that survived infection were protected from rechallenge without additional treatment. In immune-deficient mice, BCV extended survival. The data show that BCV controls viral replication at the site of challenge and reduces viral dissemination to internal organs, thus providing a shield for the developing adaptive immunity that clears the host of virus and builds virus-specific immunological memory.


Subject(s)
Antiviral Agents/administration & dosage , Cytosine/analogs & derivatives , Organophosphonates/administration & dosage , Protective Agents/administration & dosage , T-Lymphocytes/cytology , Vaccinia virus/drug effects , Vaccinia/drug therapy , Animals , Cytosine/administration & dosage , Female , Humans , Luciferases/genetics , Luciferases/metabolism , Lymphocyte Count , Mice , Mice, Inbred BALB C , Mice, Nude , T-Lymphocytes/immunology , Vaccinia/immunology , Vaccinia/mortality , Vaccinia/virology , Vaccinia virus/genetics , Vaccinia virus/physiology , Viral Load/drug effects
9.
PLoS One ; 9(6): e100547, 2014.
Article in English | MEDLINE | ID: mdl-24945301

ABSTRACT

BACKGROUND: High-dose synchrotron microbeam radiation therapy (MRT) has shown the potential to deliver improved outcomes over conventional broadbeam (BB) radiation therapy. To implement synchrotron MRT clinically for cancer treatment, it is necessary to undertake dose equivalence studies to identify MRT doses that give similar outcomes to BB treatments. AIM: To develop an in vitro approach to determine biological dose equivalence between MRT and BB using two different cell-based assays. METHODS: The acute response of tumour and normal cell lines (EMT6.5, 4T1.2, NMuMG, EMT6.5ch, 4T1ch5, SaOS-2) to MRT (50-560 Gy) and BB (1.5-10 Gy) irradiation was investigated using clonogenic and real time cell impedance sensing (RT-CIS)/xCELLigence assays. MRT was performed using a lattice of 25 or 50 µm-wide planar, polychromatic kilovoltage X-ray microbeams with 200 µm peak separation. BB irradiations were performed using a Co60 teletherapy unit or a synchrotron radiation source. BB doses that would generate biological responses similar to MRT were calculated by data interpolation and verified by clonogenic and RT-CIS assays. RESULTS: For a given cell line, MRT equivalent BB doses identified by RT-CIS/xCELLigence were similar to those identified by clonogenic assays. Dose equivalence between MRT and BB were verified in vitro in two cell lines; EMT6.5ch and SaOS-2 by clonogenic assays and RT-CIS/xCELLigence. We found for example, that BB doses of 3.4±0.1 Gy and 4.40±0.04 Gy were radiobiologically equivalent to a peak, microbeam dose of 112 Gy using clonogenic and RT-CIS assays respectively on EMT6.5ch cells. CONCLUSION: Our data provides the first determination of biological dose equivalence between BB and MRT modalities for different cell lines and identifies RT-CIS/xCELLigence assays as a suitable substitute for clonogenic assays. These results will be useful for the safe selection of MRT doses for future veterinary and clinical trials.


Subject(s)
Biological Assay/methods , Synchrotrons , Animals , Australia , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Clone Cells , Dose-Response Relationship, Radiation , Electric Impedance , Mice , Therapeutic Equivalency
10.
PLoS One ; 9(5): e98517, 2014.
Article in English | MEDLINE | ID: mdl-24870145

ABSTRACT

Prostaglandin E2 (PGE2) is induced in vivo by bacterial products including TLR agonists. To determine whether PGE2 is induced directly or via IL-1ß, human monocytes and macrophages were cultured with LPS or with Pam3CSK4 in presence of caspase-1 inhibitor, ZVAD, or IL-1R antagonist, Kineret. TLR agonists induced PGE2 in macrophages exclusively via IL-1ß-independent mechanisms. In contrast, ZVAD and Kineret reduced PGE2 production in LPS-treated (but not in Pam3CSK4-treated) monocytes, by 30-60%. Recombinant human IL-1ß augmented COX-2 and mPGES-1 mRNA and PGE2 production in LPS-pretreated monocytes but not in un-primed or Pam3CSK4-primed monocytes. This difference was explained by the finding that LPS but not Pam3CSK4 induced phosphorylation of IRF3 in monocytes suggesting activation of the TRIF signaling pathway. Knocking down TRIF, TRAM, or IRF3 genes by siRNA inhibited IL-1ß-induced COX-2 and mPGES-1 mRNA. Blocking of TLR4 endocytosis during LPS priming prevented the increase in PGE2 production by exogenous IL-1ß. Our data showed that TLR2 agonists induce PGE2 in monocytes independently from IL-1ß. In the case of TLR4, IL-1ß augments PGE2 production in LPS-primed monocytes (but not in macrophages) through a mechanism that requires TLR4 internalization and activation of the TRIF/IRF3 pathway. These findings suggest a key role for blood monocytes in the rapid onset of fever in animals and humans exposed to bacterial products and some novel adjuvants.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Dinoprostone/biosynthesis , Fever/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Monocytes/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Cells, Cultured , Cyclooxygenase 2/metabolism , Fever/microbiology , Flow Cytometry , Humans , Interferon Regulatory Factor-3/metabolism , Interleukin 1 Receptor Antagonist Protein , Lipopeptides , Lipopolysaccharides , Mice , Mice, Inbred NOD , Oligopeptides , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , Toll-Like Receptor 2/agonists
11.
Reproduction ; 147(5): 683-92, 2014 May.
Article in English | MEDLINE | ID: mdl-24713395

ABSTRACT

Uterine fibroids are a prevalent gynaecological condition in reproductive-aged women and are the commonest reason for hysterectomy. The cellular composition of clonal fibroids are heterogeneous, with phenotypically dissimilar cells that include smooth muscle cells (SMC), vascular SMC (VSMC) and fibroblasts. The aim of our study was to investigate genes that are commonly differentially expressed between fibroid and myometrial whole tissues in phenotypically different sub-populations of cells isolated from fibroid and myometrium. Genes to be investigated by fluorescence-activated cell sorting, quantitative real-time PCR and immunocytochemistry include transforming growth factor ß (TGFB) and retinoic acid (RA) signalling families and steroid hormone receptors. We hypothesised that each cell population isolated from fibroid and myometrium would differ in the expression of fibroid-associated genes. We demonstrated that phenotypically different cellular constituents of uterine fibroids differentially express cellular RA-binding protein 2 (CRABP2), progesterone receptor B (PRB) and TGFB receptor 2 mRNA in fibroid-derived cells of VSMC and SMC phenotype. CRABP2 mRNA was also differentially expressed in fibroblasts and VSMC sub-populations from within clonal fibroid tumours. We conclude that differential regulation of RA, TGFB and PR pathway transcription occurs in fibroid-associated SMC and -fibroblasts and that investigation of paracrine interactions between different cell types within the fibroid microenvironment provides an important new paradigm for understanding the pathophysiology of this common disease.


Subject(s)
Leiomyoma/metabolism , Myometrium/metabolism , Receptors, Progesterone/metabolism , Receptors, Retinoic Acid/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Gonadal Steroid Hormones/physiology , Humans , Leiomyoma/pathology , Middle Aged , Muscle, Smooth/metabolism , Muscle, Smooth/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myometrium/pathology , Paracrine Communication/physiology , Phenotype , Receptors, Progesterone/genetics , Receptors, Retinoic Acid/genetics , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/physiology , Transforming Growth Factors/physiology , Tretinoin/physiology
12.
Mol Hum Reprod ; 20(3): 250-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24243625

ABSTRACT

Uterine fibroids are conventionally defined as clonally derived benign tumours from the proliferation of a single smooth muscle cell (SMC). We have previously identified fibroblast-like cells in fibroids, the presence of which raises the question as to whether all cells within the fibroid have the same clonal origin. The first aim of this study was to develop a fluorescence-activated cell sorting (FACS)-based method to isolate different cell types from human myometrium and fibroid tissues. Secondly, we aimed to use X chromosome inactivation analysis to determine the clonality of cell subpopulations isolated from myometrial and fibroid tissues. Human myometrium and fibroid tissues were collected from women undergoing hysterectomy. Immunohistochemistry (IHC) and flow cytometry confirmed that in addition to SMCs, fibroblasts constitute a significant proportion of cells in human myometrium and fibroid tissues. FACS based on CD90 and ALDH1 reliably separated cells into three myometrial and four fibroid subpopulations: SMCs, vascular smooth muscle cells and two fibroblast subsets. Clonality was first determined by X chromosome inactivation using the classic DNA methylation-sensitive HUMARA assay. Data from this assay were highly variable, with only a quarter of samples meeting the definition of clonal fibroid and non-clonal myometrium. However, using an RNA-based X chromosome inactivation HUMARA assay, we were able to demonstrate clonality of all cellular constituents of most fibroids. Our results confirm that most fibroids are derived from a single cell, and for the first time demonstrates that these clonal cells differentiate into fibroblast and SMC subpopulation as the fibroid grows.


Subject(s)
Fibroblasts/pathology , Leiomyoma/pathology , Myocytes, Smooth Muscle/pathology , Myometrium/pathology , Aldehyde Dehydrogenase 1 Family , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Clone Cells , Female , Fibroblasts/metabolism , Flow Cytometry , Humans , Hysterectomy , Isoenzymes/metabolism , Leiomyoma/metabolism , Leiomyoma/surgery , Middle Aged , Myocytes, Smooth Muscle/metabolism , Myometrium/metabolism , Myometrium/surgery , Retinal Dehydrogenase/metabolism , Thy-1 Antigens/metabolism , X Chromosome Inactivation
13.
Reproduction ; 146(2): 91-102, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23704310

ABSTRACT

Uterine fibroids are the most common benign tumour afflicting women of reproductive age. Despite the large healthcare burden caused by fibroids, there is only limited understanding of the molecular mechanisms that drive fibroid pathophysiology. Although a large number of genes are differentially expressed in fibroids compared with myometrium, it is likely that most of these differences are a consequence of the fibroid presence and are not causal. The aim of this study was to investigate the expression and regulation of NR2F2 and CTNNB1 based on their potential causal role in uterine fibroid pathophysiology. We used real-time quantitative RT-PCR, western blotting and immunohistochemistry to describe the expression of NR2F2 and CTNNB1 in matched human uterine fibroid and myometrial tissues. Primary myometrial and fibroid smooth muscle cell cultures were treated with progesterone and/or retinoic acid (RA) and sonic hedgehog (SHH) conditioned media to investigate regulatory pathways for these proteins. We showed that NR2F2 and CTNNB1 are aberrantly expressed in fibroid tissue compared with matched myometrium, with strong blood vessel-specific localisation. Although the SHH pathway was shown to be active in myometrial and fibroid primary cultures, it did not regulate NR2F2 or CTNNB1 mRNA expression. However, progesterone and RA combined regulated NR2F2 mRNA, but not CTNNB1, in myometrial but not fibroid primary cultures. In conclusion, we demonstrate aberrant expression and regulation of NR2F2 and CTNNB1 in uterine fibroids compared with normal myometrium, consistent with the hypothesis that these factors may play a causal role uterine fibroid development.


Subject(s)
COUP Transcription Factor II/metabolism , Gene Expression Regulation, Neoplastic , Leiomyomatosis/metabolism , Myometrium/metabolism , Neoplasm Proteins/metabolism , Uterine Neoplasms/metabolism , beta Catenin/metabolism , Adult , Blood Vessels/metabolism , Blood Vessels/pathology , COUP Transcription Factor II/genetics , Cells, Cultured , Female , Follicular Phase/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Leiomyomatosis/blood supply , Leiomyomatosis/pathology , Leiomyomatosis/surgery , Luteal Phase/metabolism , Middle Aged , Myometrium/blood supply , Myometrium/pathology , Neoplasm Proteins/genetics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Progesterone/metabolism , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Tretinoin/metabolism , Tumor Cells, Cultured , Uterine Neoplasms/blood supply , Uterine Neoplasms/pathology , beta Catenin/genetics
14.
J Virol ; 87(10): 5564-76, 2013 May.
Article in English | MEDLINE | ID: mdl-23468500

ABSTRACT

Whole-body bioimaging was used to study dissemination of vaccinia virus (VACV) in normal and in immune deficient (nu(-)/nu(-)) mice protected from lethality by postchallenge administration of ST-246. Total fluxes were recorded in the liver, spleen, lungs, and nasal cavities of live mice after intranasal infection with a recombinant IHD-J-Luc VACV expressing luciferase. Areas under the flux curve were calculated for individual mice to assess viral loads. Treatment for 2 to 5 days of normal BALB/c mice with ST-246 at 100 mg/kg starting 24 h postchallenge conferred 100% protection and reduced viral loads in four organs compared to control mice. Mice also survived after 5 days of treatment with ST-246 at 30 mg/kg, and yet the viral loads and poxes were higher in these mice compared to 100-mg/kg treatment group. Nude mice were not protected by ST-246 alone or by 10 million adoptively transferred T cells. In contrast, nude mice that received T cells and 7-day treatment with ST-246 survived infection and exhibited reduced viral loads compared to nonreconstituted and ST-246-treated mice after ST-246 was stopped. Similar protection of nude mice was achieved using adoptively transferred 1.0 and 0.1 million, but not 0.01 million, purified T cells or CD4(+) or CD8(+) T cells in conjunction with ST-246 treatment. These data suggest that ST-246 protects immunocompetent mice from lethality and reduces viral dissemination in internal organs and poxvirus lesions. Furthermore, immune-deficient animals with partial T cell reconstitution can control virus replication after a course of ST-246 and survive lethal vaccinia virus challenge.


Subject(s)
Adoptive Transfer , Antiviral Agents/administration & dosage , Benzamides/administration & dosage , Isoindoles/administration & dosage , T-Lymphocytes/immunology , Vaccinia virus/pathogenicity , Vaccinia/pathology , Vaccinia/therapy , Animal Structures/virology , Animals , Disease Models, Animal , Female , Genes, Reporter , Immunocompromised Host , Luciferases/analysis , Luciferases/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Staining and Labeling , Survival Analysis , Treatment Outcome , Viral Load , Whole Body Imaging
15.
Reprod Sci ; 20(4): 361-70, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22995988

ABSTRACT

Despite the prevalence of uterine fibroids (Fs), few studies have investigated the links between clinical features and the cellular or molecular mechanisms that drive F growth and development. Such knowledge will ultimately help to differentiate symptomatic from asymptomatic Fs and could result in the development of more effective and individualized treatments. The aim of this study was to investigate the relationship between ultrasound appearance, blood flow, and angiogenic gene expression in F, perifibroid (PM), and distant myometrial (DM) tissues. We hypothesized that angiogenic gene expression would be increased in tissues and participants that showed increased blood flow by Doppler ultrasound. The study was performed using Doppler ultrasound to measure blood flow prior to hysterectomy, with subsequent tissue samples from the F, PM, and DM being investigated for angiogenic gene expression. Overall, PM blood flow (measured as peak systolic velocity [PSV]) was higher than F blood flow, although significant heterogeneity was seen in vascularity and blood flow between different Fs and their surrounding myometrium. We did not find any correlation between PSV and any other clinical or molecular parameter in this study. We identified 19 angiogenesis pathway-related genes with significant differences in expression between F and DM, and 2 genes, matrix metalloproteinase 9 (MMP9) and Neuropilin 2 (NRP2), that were significantly different between F and PM. These results are consistent with subtle differences between PM and DM. Understanding the differences between symptomatic versus asymptomatic Fs may eventually lead to more effective treatments that directly target the source of heavy menstrual bleeding.


Subject(s)
Leiomyoma/diagnostic imaging , Leiomyoma/genetics , Menorrhagia/diagnostic imaging , Menorrhagia/genetics , Transcriptome/genetics , Ultrasonography, Doppler, Color , Adult , Female , Humans , Middle Aged , Ultrasonography, Doppler, Color/methods , Uterus/blood supply , Uterus/diagnostic imaging , Uterus/physiology
16.
Radiat Res ; 178(4): 249-59, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22974124

ABSTRACT

The majority of cancer patients achieve benefit from radiotherapy. A significant limitation of radiotherapy is its relatively low therapeutic index, defined as the maximum radiation dose that causes acceptable normal tissue damage to the minimum dose required to achieve tumor control. Recently, a new radiotherapy modality using synchrotron-generated X-ray microbeam radiotherapy has been demonstrated in animal models to ablate tumors with concurrent sparing of normal tissue. Very little work has been undertaken into the cellular and molecular mechanisms that differentiate microbeam radiotherapy from broad beam. The purpose of this study was to investigate and compare the whole genome transcriptional response of in vivo microbeam radiotherapy versus broad beam irradiated tumors. We hypothesized that gene expression changes after microbeam radiotherapy are different from those seen after broad beam. We found that in EMT6.5 tumors at 4-48 h postirradiation, microbeam radiotherapy differentially regulates a number of genes, including major histocompatibility complex (MHC) class II antigen gene family members, and other immunity-related genes including Ciita, Ifng, Cxcl1, Cxcl9, Indo and Ubd when compared to broad beam. Our findings demonstrate molecular differences in the tumor response to microbeam versus broad beam irradiation and these differences provide insight into the underlying mechanisms of microbeam radiotherapy and broad beam.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic/radiation effects , Neoplasms, Experimental/radiotherapy , Synchrotrons , Animals , Cell Line, Tumor , Chemokine CXCL1/analysis , Genes, MHC Class II , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology
17.
Methods Mol Biol ; 890: 161-76, 2012.
Article in English | MEDLINE | ID: mdl-22688767

ABSTRACT

Preclinical evaluation of novel anti-smallpox vaccines and antiviral treatments often rely on mouse -challenge models using pathogenic vaccinia virus, such as Western Reserve (WR) strain or other orthopoxviruses. Traditionally, efficacy of treatment is evaluated using various readouts, such as lethality (rare), measurements of body weight loss, pox lesion scoring, and determination of viral loads in internal organs by enumerating plaques in sensitive cell lines. These methodologies provide valuable information about the contribution of the treatment to protection from infection, yet all have similar limitations: they do not evaluate dissemination of the virus within the same animal and require large numbers of animals. These two problems prompted us to turn to a recently developed whole body imaging technology, where replication of recombinant vaccinia virus expressing luciferase enzyme (WRvFire) is sensed by detecting light emitted by the enzyme in the presence of D: -luciferin substrate administered to infected animal. Bioluminescence signals from infected organs in live animals are registered by the charge-coupled device camera in IVIS instrument developed by Caliper, and are converted into numerical values. This chapter describes whole body bioimaging methodology used to determine viral loads in normal live BALB/c mice infected with recombinant WRvFire vaccinia virus. Using Dryvax vaccination as a model, we show how bioluminescence data can be used to determine efficacy of treatment. In addition, we illustrate how bioluminescence and survival outcome can be combined in Receiver Operating Characteristic curve -analysis to develop predictive models of lethality that can be applied for testing of new therapeutics and second-generation vaccines.


Subject(s)
Antiviral Agents/therapeutic use , Smallpox Vaccine/administration & dosage , Vaccinia virus/physiology , Vaccinia/pathology , Animals , Antigens, Viral/administration & dosage , Antiviral Agents/pharmacology , Area Under Curve , Drug Evaluation, Preclinical/methods , Firefly Luciferin/administration & dosage , Genes, Reporter , Liver/virology , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Luminescent Measurements , Lung/virology , Mice , Mice, Inbred BALB C , Nasal Cavity/virology , ROC Curve , Spleen/virology , Vaccination , Vaccinia/drug therapy , Vaccinia/prevention & control , Vaccinia virus/immunology , Vaccinia virus/metabolism , Whole Body Imaging
18.
Vaccine ; 30(32): 4859-65, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22609036

ABSTRACT

Subunit vaccines composed of recombinant or purified antigens have a good safety record but are poorly immunogenic and require adjuvants to activate innate immunity and facilitate antigen specific immune response. Of the many adjuvant formulations that are under development, very few are licensed mainly due to concerns about adverse side effects. The goal of our study was to develop in vitro assays that could predict toxicity of adjuvants in vivo. Pro-inflammatory cytokines IL-ß, IL-6, TNF-α, and IL-8 were measured in human primary monocytes and the monocytoid cell line, MonoMac 6 (MM6), activated with a panel of TLR agonists or with adjuvants. A 0.5 EU/ml dose of Standard for endotoxin (previously shown to provide a margin between pyrogenic and non-pyrogenic substances in rabbits) was used as a comparator to establish a "safety threshold". FSL-1, Pam3CSK4, flagellin, and R848 TLR agonists but not Alum, MF59, Poly I:C, or MPL adjuvants induced cytokines in MM6 cells above the safety threshold. To confirm the predictive value of the in vitro assays, FSL-1 and flagellin were injected intramuscularly into New Zealand White (NZW) rabbits. Both TLR agonists induced fever within 6-8h post-injection followed 24-48 h later by increased C reactive protein (CRP). Importantly, an early peak in plasma prostaglandin E2 (PGE(2)) levels preceded rise in body temperature. In vitro production of PGE(2) in monocytes and MM6 cells was found following treatments with various TLR agonists but not with alum, MF59, MPL, or Poly I:C adjuvants. Together, our studies demonstrated a strong correlation between production of pro-inflammatory cytokines above a "safety threshold" and production of PGE(2)in vitro and an increase in body temperature in rabbits. The developed human cell based assays could provide an important tool for early screening of new molecular moieties and adjuvant formulations and may assist in selection of safer products.


Subject(s)
Adjuvants, Immunologic/pharmacology , Monocytes/drug effects , Toll-Like Receptors/agonists , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/standards , Animals , Biological Assay , C-Reactive Protein/analysis , Cell Line , Cytokines/analysis , Dinoprostone/analysis , Female , Fever/chemically induced , Humans , Predictive Value of Tests , Rabbits , Reference Standards
19.
Virol J ; 8: 441, 2011 Sep 20.
Article in English | MEDLINE | ID: mdl-21933385

ABSTRACT

BACKGROUND: Previously we demonstrated that DNA vaccination of nonhuman primates (NHP) with a small subset of vaccinia virus (VACV) immunogens (L1, A27, A33, B5) protects against lethal monkeypox virus challenge. The L1 and A27 components of this vaccine target the mature virion (MV) whereas A33 and B5 target the enveloped virion (EV). RESULTS: Here, we demonstrated that the antibodies produced in vaccinated NHPs were sufficient to confer protection in a murine model of lethal Orthopoxvirus infection. We further explored the concept of using DNA vaccine technology to produce immunogen-specific polyclonal antibodies that could then be combined into cocktails as potential immunoprophylactic/therapeutics. Specifically, we used DNA vaccines delivered by muscle electroporation to produce polyclonal antibodies against the L1, A27, A33, and B5 in New Zealand white rabbits. The polyclonal antibodies neutralized both MV and EV in cell culture. The ability of antibody cocktails consisting of anti-MV, anti-EV, or a combination of anti-MV/EV to protect BALB/c mice was evaluated as was the efficacy of the anti-MV/EV mixture in a mouse model of progressive vaccinia. In addition to evaluating weight loss and lethality, bioimaging technology was used to characterize the spread of the VACV infections in mice. We found that the anti-EV cocktail, but not the anti-MV cocktail, limited virus spread and lethality. CONCLUSIONS: A combination of anti-MV/EV antibodies was significantly more protective than anti-EV antibodies alone. These data suggest that DNA vaccine technology could be used to produce a polyclonal antibody cocktail as a possible product to replace vaccinia immune globulin.


Subject(s)
Antibodies, Viral/administration & dosage , Antigens, Viral/immunology , Smallpox Vaccine/genetics , Vaccines, DNA/genetics , Vaccinia virus/immunology , Vaccinia/prevention & control , Viral Envelope Proteins/genetics , Animals , Antibodies, Heterophile , Antigens, Viral/chemistry , Antigens, Viral/genetics , Body Weight , Disease Models, Animal , Electroporation , Female , Mice , Mice, Inbred BALB C , Molecular Imaging , Muscles/immunology , Neutralization Tests , Rabbits , Smallpox Vaccine/immunology , Vaccines, DNA/immunology , Vaccinia/immunology , Vaccinia virus/drug effects , Vaccinia virus/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology , Virion/drug effects , Virion/immunology
20.
J Virol ; 85(17): 9147-58, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21715493

ABSTRACT

Whole-body bioimaging was employed to study the effects of passive immunotherapies on lethality and viral dissemination in BALB/c mice challenged with recombinant vaccinia viruses expressing luciferase. WRvFire and IHD-J-Luc vaccinia viruses induced lethality with similar times to death following intranasal infection, but WRvFire replicated at higher levels than IHD-J-Luc in the upper and lower respiratory tracts. Three types of therapies were tested: licensed human anti-vaccinia virus immunoglobulin intravenous (VIGIV); recombinant anti-vaccinia virus immunoglobulin (rVIG; Symphogen, Denmark), an investigational product containing a mixture of 26 human monoclonal antibodies (HuMAbs) against mature virion (MV) and enveloped virion (EV); and HuMAb compositions targeting subsets of MV or EV proteins. Bioluminescence recorded daily showed that pretreatment with VIGIV (30 mg) or with rVIG (100 µg) on day -2 protected mice from death but did not prevent viral replication at the site of inoculation and dissemination to internal organs. Compositions containing HuMAbs against MV or EV proteins were protective in both infection models at 100 µg per animal, but at 30 µg, only anti-EV antibodies conferred protection. Importantly, the t statistic of the mean total fluxes revealed that viral loads in surviving mice were significantly reduced in at least 3 sites for 3 consecutive days (days 3 to 5) postchallenge, while significant reduction for 1 or 2 days in any individual site did not confer protection. Our data suggest that reduction of viral replication at multiple sites, including respiratory tract, spleen, and liver, as monitored by whole-body bioluminescence can be used to predict the effectiveness of passive immunotherapies in mouse models.


Subject(s)
Animal Structures/virology , Immunization, Passive/methods , Respiratory System/virology , Vaccinia virus/pathogenicity , Vaccinia/mortality , Vaccinia/prevention & control , Viral Load , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Viral/administration & dosage , Disease Models, Animal , Female , Genes, Reporter , Immunoglobulin G/administration & dosage , Luciferases/metabolism , Luminescent Measurements , Mice , Mice, Inbred BALB C , Recombinant Proteins/administration & dosage , Rodent Diseases/mortality , Rodent Diseases/prevention & control , Staining and Labeling/methods , Survival Analysis , Time Factors , Vaccinia virus/immunology , Whole Body Imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...