Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Microbiol Spectr ; 12(6): e0400523, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38709045

ABSTRACT

Clostridioides difficile infection (CDI) with high morbidity and high mortality is an urgent threat to public health, and C. difficile pathogenesis studies are eagerly required for CDI therapy. The major surface layer protein, SlpA, was supposed to play a key role in C. difficile pathogenesis; however, a lack of isogenic slpA mutants has greatly hampered analysis of SlpA functions. In this study, the whole slpA gene was successfully deleted for the first time via CRISPR-Cas9 system. Deletion of slpA in C. difficile resulted in smaller, smother-edged colonies, shorter bacterial cell size, and aggregation in suspension. For life cycle, the mutant demonstrated lower growth (changes of optical density at 600 nm, OD600) but higher cell density (colony-forming unit, CFU), decreased toxins production, and inhibited sporulation. Moreover, the mutant was more impaired in motility, more sensitive to vancomycin and Triton X-100-induced autolysis, releasing more lactate dehydrogenase. In addition, SlpA deficiency led to robust biofilm formation but weak adhesion to human host cells.IMPORTANCEClostridioides difficile infection (CDI) has been the most common hospital-acquired infection, with a high rate of antibiotic resistance and recurrence incidences, become a debilitating public health threat. It is urgently needed to study C. difficile pathogenesis for developing efficient strategies as CDI therapy. SlpA was indicated to play a key role in C. difficile pathogenesis. However, analysis of SlpA functions was hampered due to lack of isogenic slpA mutants. Surprisingly, the first slpA deletion C. difficile strain was generated in this study via CRISPR-Cas9, further negating the previous thought about slpA being essential. Results in this study will provide direct proof for roles of SlpA in C. difficile pathogenesis, which will facilitate future investigations for new targets as vaccines, new therapeutic agents, and intervention strategies in combating CDI.


Subject(s)
Bacterial Proteins , Biofilms , Clostridioides difficile , Clostridium Infections , Gene Deletion , Clostridioides difficile/genetics , Clostridioides difficile/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Clostridium Infections/microbiology , Biofilms/growth & development , Anti-Bacterial Agents/pharmacology , Virulence/genetics , CRISPR-Cas Systems , Bacterial Adhesion/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism
2.
Sci Rep ; 14(1): 11179, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38750069

ABSTRACT

During a SARS-CoV-2 infection, macrophages recognize viral components resulting in cytokine production. While this response fuels virus elimination, overexpression of cytokines can lead to severe COVID-19. Previous studies suggest that the spike protein (S) of SARS-CoV-2 can elicit cytokine production via the transcription factor NF-κB and the toll-like receptors (TLRs). In this study, we found that: (i) S and the S2 subunit induce CXCL10, a chemokine implicated in severe COVID-19, gene expression by human macrophage cells (THP-1); (ii) a glycogen synthase kinase-3 inhibitor attenuates this induction; (iii) S and S2 do not activate NF-κB but do activate the transcription factor IRF; (iv) S and S2 do not require TLR2 to elicit CXCL10 production or activate IRF; and (v) S and S2 elicit CXCL10 production by peripheral blood mononuclear cells (PBMCs). We also discovered that the cellular response, or lack thereof, to S and S2 is a function of the recombinant S and S2 used. While such a finding raises the possibility of confounding LPS contamination, we offer evidence that potential contaminating LPS does not underly induced increases in CXCL10. Combined, these results provide insights into the complex immune response to SARS-CoV-2 and suggest possible therapeutic targets for severe COVID-19.


Subject(s)
COVID-19 , Chemokine CXCL10 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Chemokine CXCL10/metabolism , Humans , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/immunology , COVID-19/virology , COVID-19/immunology , COVID-19/metabolism , Macrophages/metabolism , Macrophages/immunology , Macrophages/virology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , NF-kappa B/metabolism , THP-1 Cells
3.
J Inflamm Res ; 16: 5339-5366, 2023.
Article in English | MEDLINE | ID: mdl-38026235

ABSTRACT

Purpose: Non-alcoholic fatty liver disease (NAFLD), recently renamed metabolic (dysfunction) associated fatty liver disease (MAFLD), is the most common chronic liver disease in the United States. Presently, there is an intense and ongoing effort to identify and develop novel therapeutics for this disease. In this study, we explored the anti-inflammatory activity of a new compound, termed IOI-214, and its therapeutic potential to ameliorate NAFLD/MAFLD in male C57BL/6J mice fed a high fat (HF) diet. Methods: Murine macrophages and hepatocytes in culture were treated with lipopolysaccharide (LPS) ± IOI-214 or DMSO (vehicle), and RT-qPCR analyses of inflammatory cytokine gene expression were used to assess IOI-214's anti-inflammatory properties in vitro. Male C57BL/6J mice were also placed on a HF diet and treated once daily with IOI-214 or DMSO for 16 weeks. Tissues were collected and analyzed to determine the effects of IOI-214 on HF diet-induced NAFL D/MAFLD. Measurements such as weight, blood glucose, serum cholesterol, liver/serum triglyceride, insulin, and glucose tolerance tests, ELISAs, metabolomics, Western blots, histology, gut microbiome, and serum LPS binding protein analyses were conducted. Results: IOI-214 inhibited LPS-induced inflammation in macrophages and hepatocytes in culture and abrogated HF diet-induced mesenteric fat accumulation, hepatic inflammation and steatosis/hepatocellular ballooning, as well as fasting hyperglycemia without affecting insulin resistance or fasting insulin, cholesterol or TG levels despite overall obesity in vivo in male C57BL/6J mice. IOI-214 also decreased systemic inflammation in vivo and improved gut microbiota dysbiosis and leaky gut. Conclusion: Combined, these data indicate that IOI-214 works at multiple levels in parallel to inhibit the inflammation that drives HF diet-induced NAFLD/MAFLD, suggesting that it may have therapeutic potential for NAFLD/MAFLD.

4.
Biochem Biophys Res Commun ; 605: 171-176, 2022 05 21.
Article in English | MEDLINE | ID: mdl-35367865

ABSTRACT

A key component of severe COVID-19 is a "cytokine storm" i.e., the excessive expression of unneeded cytokines. Previous studies suggest that SARS-CoV-2 proteins can induce macrophages to secrete pro-inflammatory cytokines; a process that may involve Toll-like receptors (TLRs). Glycogen synthase kinase-3 (GSK-3) has been implicated in TLR signal transduction and a selective GSK-3 inhibitor, termed COB-187, dramatically attenuates cytokine expression induced by the TLR ligand lipopolysaccharide (LPS). In the present study, we provide evidence that the SARS-CoV-2 spike protein (S) and the S2 subunit (S2) induce production of CXCL10 (a chemokine elevated in severe COVID-19) by a human macrophage cell line. Further, we report that two clinically relevant GSK-3 inhibitors and COB-187 attenuate S and S2 protein-induced CXCL10 production. Combined, our observations provide impetus for investigating GSK-3 inhibitors as potential therapeutics for severe COVID-19.


Subject(s)
COVID-19 Drug Treatment , Glycogen Synthase Kinase 3 , Cytokines/metabolism , Humans , SARS-CoV-2 , Spike Glycoprotein, Coronavirus
5.
Pancreas ; 51(1): 48-55, 2022 01 01.
Article in English | MEDLINE | ID: mdl-35195595

ABSTRACT

OBJECTIVE: Genetic and environmental influences play a role as triggers of type 1 diabetes mellitus (T1DM). Female nonobese diabetic (NOD) mice are useful for studying T1DM as they spontaneously develop T1DM, which can be accelerated by some viruses. Toll-like receptor 3 (TLR3) is believed to play a critical role in viral-induced T1DM and ß-cell destruction, because female Tlr3 knockout (Tlr3-/-) NOD mice are protected from Coxsackievirus B4 (CVB4)-induced acceleration of T1DM. However, the exact role(s) TLR3 plays in the pathogenesis of CVB4-induced T1DM remain unknown. METHODS: This longitudinal study used immunostaining, laser capture microdissection, and reverse transcription real-time polymerase chain reaction of islets from female uninfected and CVB4-infected Tlr3+/+ and Tlr3-/- NOD mice. RESULTS: Islets isolated from female Tlr3+/+ NOD mice 4 to 8 weeks of age had higher amounts of insulitis, Cxcl10, Il1b, Tnfa, and Tgfb1 expression compared with Tlr3-/- NOD mice. After CVB4 infection, Tlr3+/+ NOD mice had higher amounts of insulitis and T-cell infiltration at 3 days after infection compared with Tlr3-/- CVB4-infected NOD mice. CONCLUSIONS: Toll-like receptor 3 is necessary for establishment of a pancreatic islet inflammatory microenvironment by increasing insulitis and cytokine expression that facilitates CVB4-induced T1DM in female NOD mice.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/chemically induced , Islets of Langerhans/metabolism , Receptors, Virus/metabolism , Toll-Like Receptor 3/metabolism , Animals , Female , Immunochemistry , Longitudinal Studies , Mice , Mice, Inbred NOD
6.
Eur J Pharmacol ; 883: 173340, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32634441

ABSTRACT

Sepsis is a serious condition that can lead to long-term organ damage and death. At the molecular level, the hallmark of sepsis is the elevated expression of a multitude of potent cytokines, i.e. a cytokine storm. For sepsis involving gram-negative bacteria, macrophages recognize lipopolysaccharide (LPS) shed from the bacteria, activating Toll-like-receptor 4 (TLR4), and triggering a cytokine storm. Glycogen synthase kinase-3 (GSK-3) is a highly active kinase that has been implicated in LPS-induced cytokine production. Thus, compounds that inhibit GSK-3 could be potential therapeutics for sepsis. Our group has recently described a novel and highly selective inhibitor of GSK-3 termed COB-187. In the present study, using THP-1 macrophages, we evaluated the ability of COB-187 to attenuate LPS-induced cytokine production. We found that COB-187 significantly reduced, at the protein and mRNA levels, cytokines induced by LPS (e.g. IL-6, TNF-α, IL-1ß, CXCL10, and IFN-ß). Further, the data suggest that the inhibition could be due, at least in part, to COB-187 reducing NF-κB (p65/p50) DNA binding activity as well as reducing IRF-3 phosphorylation at Serine 396. Thus, COB-187 appears to be a potent inhibitor of the cytokine storm induced by LPS.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokine Release Syndrome/prevention & control , Cytokines/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Inflammation Mediators/metabolism , Macrophages/drug effects , Protein Kinase Inhibitors/pharmacology , Cytokine Release Syndrome/chemically induced , Cytokine Release Syndrome/enzymology , Cytokine Release Syndrome/genetics , Cytokines/genetics , Down-Regulation , Glycogen Synthase Kinase 3/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Lipopolysaccharides/toxicity , Macrophages/enzymology , NF-kappa B/metabolism , Phosphorylation , Sepsis/chemically induced , Sepsis/enzymology , Sepsis/genetics , Sepsis/prevention & control , Signal Transduction , THP-1 Cells
7.
Am J Physiol Cell Physiol ; 317(6): C1289-C1303, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31553649

ABSTRACT

Glycogen synthase kinase-3 (GSK-3) is a multitasking protein kinase that regulates numerous critical cellular functions. Not surprisingly, elevated GSK-3 activity has been implicated in a host of diseases including pathological inflammation, diabetes, cancer, arthritis, asthma, bipolar disorder, and Alzheimer's. Therefore, reagents that inhibit GSK-3 activity provide a means to investigate the role of GSK-3 in cellular physiology and pathophysiology and could become valuable therapeutics. Finding a potent inhibitor of GSK-3 that can selectively target this kinase, among over 500 protein kinases in the human genome, is a significant challenge. Thus there remains a critical need for the identification of selective inhibitors of GSK-3. In this work, we introduce a novel small organic compound, namely COB-187, which exhibits potent and highly selective inhibition of GSK-3. Specifically, this study 1) utilized a molecular screen of 414 kinase assays, representing 404 unique kinases, to reveal that COB-187 is a highly potent and selective inhibitor of GSK-3; 2) utilized a cellular assay to reveal that COB-187 decreases the phosphorylation of canonical GSK-3 substrates indicating that COB-187 inhibits cellular GSK-3 activity; and 3) reveals that a close isomer of COB-187 is also a selective and potent inhibitor of GSK-3. Taken together, these results demonstrate that we have discovered a region of chemical design space that contains novel GSK-3 inhibitors. These inhibitors will help to elucidate the intricate function of GSK-3 and can serve as a starting point for the development of potential therapeutics for diseases that involve aberrant GSK-3 activity.


Subject(s)
Biphenyl Compounds/pharmacology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Animals , Biphenyl Compounds/chemical synthesis , Drug Design , Enzyme Assays , Gene Expression , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , HEK293 Cells , High-Throughput Screening Assays , Humans , Mice , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Protein Kinases/genetics , RAW 264.7 Cells , Structure-Activity Relationship , Substrate Specificity , THP-1 Cells , Tetradecanoylphorbol Acetate/pharmacology , Thiadiazoles/chemistry , Thiadiazoles/pharmacology
8.
Endocrinology ; 156(2): 453-61, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25422874

ABSTRACT

Group B coxsackieviruses (CVBs) are involved in triggering some cases of type 1 diabetes mellitus (T1DM). However, the molecular mechanism(s) responsible for this remain elusive. Toll-like receptor 3 (TLR3), a receptor that recognizes viral double-stranded RNA, is hypothesized to play a role in virus-induced T1DM, although this hypothesis is yet to be substantiated. The objective of this study was to directly investigate the role of TLR3 in CVB-triggered T1DM in nonobese diabetic (NOD) mice, a mouse model of human T1DM that is widely used to study both spontaneous autoimmune and viral-induced T1DM. As such, we infected female wild-type (TLR3(+/+)) and TLR3 knockout (TLR3(-/-)) NOD mice with CVB4 and compared the incidence of diabetes in CVB4-infected mice with that of uninfected counterparts. We also evaluated the islets of uninfected and CVB4-infected wild-type and TLR3 knockout NOD mice by immunohistochemistry and insulitis scoring. TLR3 knockout mice were markedly protected from CVB4-induced diabetes compared with CVB4-infected wild-type mice. CVB4-induced T-lymphocyte-mediated insulitis was also significantly less severe in TLR3 knockout mice compared with wild-type mice. No differences in insulitis were observed between uninfected animals, either wild-type or TLR3 knockout mice. These data demonstrate for the first time that TLR3 is 1) critical for CVB4-induced T1DM, and 2) modulates CVB4-induced insulitis in genetically prone NOD mice.


Subject(s)
Coxsackievirus Infections/complications , Diabetes Mellitus, Type 1/virology , Toll-Like Receptor 3/metabolism , Animals , Coxsackievirus Infections/metabolism , Diabetes Mellitus, Type 1/metabolism , Enterovirus B, Human/isolation & purification , Female , Mice, Inbred NOD , Mice, Knockout , Pancreas/virology , Random Allocation
9.
Exp Cell Res ; 323(1): 7-27, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24569142

ABSTRACT

Dendritic cells (DCs) are immune cells found in the peripheral tissues where they sample the organism for infections or malignancies. There they take up antigens and migrate towards immunological organs to contact and activate T lymphocytes that specifically recognize the antigen presented by these antigen presenting cells. In the steady state there are several types of resident DCs present in various different organs. For example, in the mouse, splenic DC populations characterized by the co-expression of CD11c and CD8 surface markers are specialized in cross-presentation to CD8 T cells, while CD11c/SIRP-1α DCs seem to be dedicated to activating CD4 T cells. On the other hand, DCs have also been associated with the development of various diseases such as cancer, atherosclerosis, or inflammatory conditions. In such disease, DCs can participate by inducing angiogenesis or immunosuppression (tumors), promoting autoimmune responses, or exacerbating inflammation (atherosclerosis). This change in DC biology can be prompted by signals in the microenvironment. We have previously shown that the interaction of DCs with various extracellular matrix components modifies the immune properties and angiogenic potential of these cells. Building on those studies, herewith we analyzed the angiogenic profile of murine myeloid DCs upon interaction with 2D and 3D type-I collagen environments. As determined by PCR array technology and quantitative PCR analysis we observed that interaction with these collagen environments induced the expression of particular angiogenic molecules. In addition, DCs cultured on collagen environments specifically upregulated the expression of CXCL-1 and -2 chemokines. We were also able to establish DC cultures on type-IV collagen environments, a collagen type expressed in pathological conditions such as atherosclerosis. When we examined DC populations in atherosclerotic veins of Apolipoprotein E deficient mice we observed that they expressed adhesion molecules capable of interacting with collagen. Finally, to further investigate the interaction of DCs with collagen in other pathological conditions, we determined that both murine ovarian and breast cancer cells express several collagen molecules that can contribute to shape their particular tumor microenvironment. Consistently, tumor-associated DCs were shown to express adhesion molecules capable of interacting with collagen molecules as determined by flow cytometry analysis. Of particular relevance, tumor-associated DCs expressed high levels of CD305/LAIR-1, an immunosuppressive receptor. This suggests that signaling through this molecule upon interaction with collagen produced by tumor cells might help define the poorly immunogenic status of these cells in the tumor microenvironment. Overall, these studies demonstrate that through interaction with collagen proteins, DCs can be capable of modifying the microenvironments of inflammatory disease such as cancer or atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Breast Neoplasms/metabolism , Dendritic Cells/metabolism , Ovarian Neoplasms/metabolism , Receptors, Collagen/metabolism , Animals , Apolipoproteins E/genetics , Atherosclerosis/immunology , Breast Neoplasms/immunology , CD11c Antigen/metabolism , Cell Adhesion Molecules/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Proliferation , Chemokine CXCL1/biosynthesis , Chemokine CXCL2/biosynthesis , Chemotaxis , Collagen/metabolism , Female , Integrin alpha1beta1/biosynthesis , Integrin alpha1beta1/metabolism , Integrin alpha2beta1/biosynthesis , Integrin alpha2beta1/metabolism , Integrin alpha3beta1/biosynthesis , Integrin alpha3beta1/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Neovascularization, Physiologic , Ovarian Neoplasms/immunology , Receptors, Collagen/biosynthesis , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/metabolism , Scavenger Receptors, Class A/biosynthesis , Scavenger Receptors, Class A/metabolism , Tumor Microenvironment , Up-Regulation
10.
Molecules ; 18(4): 3841-58, 2013 Mar 27.
Article in English | MEDLINE | ID: mdl-23535518

ABSTRACT

Accumulating evidence supports a role for viruses in the pathogenesis of type 1 diabetes mellitus (T1DM). Activation of dsRNA-sensing pathways by viral dsRNA induces the production of inflammatory cytokines and chemokines that trigger beta cell apoptosis, insulitis, and autoimmune-mediated beta cell destruction. This study was designed to evaluate and describe potential protective effects of phenylmethimazole (C10), a small molecule which blocks dsRNA-mediated signaling, on preventing dsRNA activation of beta cell apoptosis and the inflammatory pathways important in the pathogenesis of T1DM. We first investigated the biological effects of C10, on dsRNA-treated pancreatic beta cells in culture. Cell viability assays, quantitative real-time PCR, and ELISAs were utilized to evaluate the effects of C10 on dsRNA-induced beta cell cytotoxicity and cytokine/chemokine production in murine pancreatic beta cells in culture. We found that C10 significantly impairs dsRNA-induced beta cell cytotoxicity and up-regulation of cytokines and chemokines involved in the pathogenesis of T1DM, which prompted us to evaluate C10 effects on viral acceleration of T1DM in NOD mice. C10 significantly inhibited viral acceleration of T1DM in NOD mice. These findings demonstrate that C10 (1) possesses novel beta cell protective activity which may have potential clinical relevance in T1DM and (2) may be a useful tool in achieving a better understanding of the role that dsRNA-mediated responses play in the pathogenesis of T1DM.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Enterovirus/pathogenicity , Insulin-Secreting Cells/drug effects , Methimazole/analogs & derivatives , RNA, Double-Stranded/adverse effects , Thiones/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Cytokines/blood , Diabetes Mellitus, Type 1/virology , Enterovirus/metabolism , Female , Inflammation/drug therapy , Inflammation/pathology , Methimazole/pharmacology , Mice , Mice, Inbred NOD , Real-Time Polymerase Chain Reaction , Signal Transduction , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Up-Regulation
11.
Immunobiology ; 218(1): 64-75, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22551928

ABSTRACT

Dendritic cells (DCs) are antigen presenting cells capable of inducing specific immune responses against microbial infections, transplant antigens, or tumors. DCs have been shown to possess a high plasticity showing different phenotypes in response to their microenvironment. For example, tumor-associated DCs can acquire an angiogenic phenotype thus promoting tumor growth. Further, DCs cultured in vitro under different conditions are able to upregulate the expression of endothelial markers and to express angiogenic factors. Indeed, it has been shown that soluble factors such as VEGF of PGE-2, that are present in the microenvironment of several tumors, affect the biology of these cells. We hypothesize that in addition to soluble factors the adhesion to different substrates will also define the phenotype and function of DCs. Herewith we demonstrate that murine myeloid(m) DCs upregulate endothelial markers such as VE-Cadherin, and to a lesser extent TIE-2, and decrease their immune capabilities when cultured on solid surfaces as compared with the same cells cultured on ultra-low binding (ULB) surfaces. On the other hand, the expression of angiogenic molecules at the level of RNA was not different among these cultures. In order to further investigate this phenomenon we used the murine ID8 model of ovarian cancer which can generate solid tumors when cancer cells are injected subcutaneously or a malignant ascites when they are injected intraperitoneally. This model gave us the unique opportunity to investigate DCs in suspension or attached to solid surfaces under the influence of the same tumor cells. We were able to determine that DCs present in solid tumors showed higher levels of expression of endothelial markers and angiogenic molecules but were not able to respond to inflammatory stimuli at the same extent as DCs recovered from ascites. Moreover, mDCs cultured on ULB surfaces in the presence of tumor factors do not expressed endothelial markers. Taking into account all these data we consider that tumor factors might be responsible for inducing angiogenic properties in DCs, but that in some settings the expression of endothelial markers such as VE-Cadherin and TIE-2 might be a function of attachment to solid surfaces and independent of the angiogenic properties of these cells.


Subject(s)
Dendritic Cells/immunology , Endothelium/metabolism , Ovarian Neoplasms/immunology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Adhesion/immunology , Cell Differentiation , Cell Line, Tumor , Endothelium/immunology , Female , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Tumor Microenvironment , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
12.
Molecules ; 17(10): 12365-77, 2012 Oct 22.
Article in English | MEDLINE | ID: mdl-23090018

ABSTRACT

Previous studies revealed that phenylmethimazole (C10) inhibits IRF3 signaling, preventing dsRNA-induction of type 1 interferon gene expression, production, and downstream signaling. In the present study, we investigated the molecular basis for C10 inhibition of dsRNA-stimulated IRF3 signaling. IRF-3 Trans-AM assays were used to measure C10 effects on dsRNA induction of IRF3 DNA binding. Green fluorescent protein-labeled IRF3 was used to measure C10 effects on dsRNA-induced IRF3 nuclear translocation. Native PAGE, SDS PAGE, and western blotting were used to identify effects of C10 on IRF3 homodimer formation and phosphorylation, respectively. There was a significant impairment of dsRNA-induced IRF3 DNA binding activity in human embryonic kidney and pancreatic cancer cells with C10 treatment. C10 also blocked dsRNA-induced IRF3 nuclear translocation and homodimer formation without blocking serine 396 phosphorylation of IRF3. Together, these results indicate that C10 interferes with IRF3 signaling by blocking dsRNA-induced IRF3 homodimer formation, a prerequisite for nuclear translocation and DNA binding activities.


Subject(s)
Cell Nucleus/metabolism , Interferon Regulatory Factor-3/metabolism , Methimazole/analogs & derivatives , Protein Multimerization/drug effects , RNA, Double-Stranded/pharmacology , Thiones/pharmacology , Cell Line, Tumor , Cell Nucleus/drug effects , DNA/metabolism , HEK293 Cells , Humans , Methimazole/pharmacology , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Transport/drug effects
13.
BMC Immunol ; 12: 35, 2011 Jun 06.
Article in English | MEDLINE | ID: mdl-21645356

ABSTRACT

BACKGROUND: Dendritic cells (DCs) are antigen presenting cells capable of inducing specific immune responses against microbial infections, transplant antigens, or tumors. Interestingly, microenvironment conditions such as those present in tumor settings might induce a DC phenotype that is poorly immunogenic and with the capability of promoting angiogenesis. We hypothesize that this plasticity may be caused not only by the action of specific cytokines or growth factors but also by the properties of the surfaces with which they interact, such as extracellular matrix (ECM) components. RESULTS: Herewith we studied the effect of different surfaces and soluble factors on the biology of DCs. To accomplish this, we cultured murine myeloid(m) DCs on surfaces coated with fibronectin, collagen I, gelatin, and Matrigel using poly-D-lysine and polystyrene as non-biological surfaces. Further, we cultured these cells in the presence of regular DC medium (RPMI 10% FBS) or commercially available endothelial medium (EGM-2). We determined that mDCs could be kept in culture up to 3 weeks in these conditions, but only in the presence of GM-CSF. We were able to determine that long-term DC cultures produce an array of angiogenic factors, and that some of these cultures still retain the capability to induce T cell responses. CONCLUSIONS: Altogether these data indicate that in order to design DC-based vaccines or treatments focused on changing the phenotype of DCs associated with diseases such as cancer or atherosclerosis, it becomes necessary to fully investigate the microenvironment in which these cells are present or will be delivered.


Subject(s)
Dendritic Cells/physiology , Myeloid Cells/physiology , Angiogenic Proteins/metabolism , Animals , Cell Culture Techniques/methods , Cell Line, Tumor , Collagen , Collagen Type I/pharmacology , Dendritic Cells/drug effects , Drug Combinations , Female , Fibronectins/pharmacology , Gelatin/pharmacology , Laminin , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Myeloid Cells/drug effects , Polylysine , Polystyrenes , Proteoglycans , T-Lymphocytes/immunology
14.
Cancer Biol Ther ; 7(8): 1194-205, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18458533

ABSTRACT

We have previously shown that intratumor administration of HSV-1716 (an ICP34.5 null mutant) resulted in significant reduction of tumor growth and a significant survival advantage in a murine model of ovarian cancer. Herewith we report that oncolytic HSV-1716 generates vaccination effects in the same model. Upon HSV-1716 infection, mouse ovarian tumor cells showed high levels of expression viral glycoproteins B and D and were highly phagocyted by dendritic cells (DCs). Interestingly, increased phagocytosis of tumor-infected cells by DCs was impaired by heparin, and anti-HSV glycoproteins B and D, indicating that viral infection enhances adhesive interactions between DCs and tumor apoptotic bodies. Moreover, HSV-1716 infected cells expressed high levels of heat shock proteins 70 and GRP94, molecules that have been reported to induce maturation of DCs, increase cross-presentation of antigens and promote antitumor immune response. After phagocytosis of tumor-infected cells, DCs acquired a mature status in vitro and in vivo, upregulated the expression of costimulatory molecule and increased migration towards MIP-3beta. Furthermore, HSV-1716 oncolytic treatment markedly reduced vascular endothelial growth factor (VEGF) levels in tumor-bearing animals thus abrogating tumor immunosuppressive milieu. These mechanisms may account for the highly enhanced antitumoral immune responses observed in HSV-1716 treated animals. Oncolytic treatment induced a significantly higher frequency of tumor-reactive IFNgamma producing cells, and induced a robust tumor infiltration by T cells. These results indicate that oncolytic therapy with HSV-1716 facilitates antitumor immune responses.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Neoplasms/immunology , Oncolytic Virotherapy/methods , Simplexvirus/immunology , Animals , Antigen Presentation/genetics , Antigens, Neoplasm/genetics , Female , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Neoplasms/virology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Ovarian Neoplasms/virology , Simplexvirus/genetics , Tumor Cells, Cultured
15.
J Transl Med ; 6: 21, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18445282

ABSTRACT

Because of the lack of full characterization of tumor associated antigens for solid tumors, whole antigen use is a convenient approach to tumor vaccination. Tumor RNA and apoptotic tumor cells have been used as a source of whole tumor antigen to prepare dendritic cell (DC) based tumor vaccines, but their efficacy has not been directly compared. Here we compare directly RNA electroporation and pulsing of DCs with whole tumor cells killed by ultraviolet (UV) B radiation using a convenient tumor model expressing human papilloma virus (HPV) E6 and E7 oncogenes. Although both approaches led to DCs presenting tumor antigen, electroporation with tumor cell total RNA induced a significantly higher frequency of tumor-reactive IFN-gamma secreting T cells, and E7-specific CD8+ lymphocytes compared to pulsing with UV-irradiated tumor cells. DCs electroporated with tumor cell RNA induced a larger tumor infiltration by T cells and produced a significantly stronger delay in tumor growth compared to DCs pulsed with UV-irradiated tumor cells. We conclude that electroporation with whole tumor cell RNA and pulsing with UV-irradiated tumor cells are both effective in eliciting antitumor immune response, but RNA electroporation results in more potent tumor vaccination under the examined experimental conditions.


Subject(s)
Cancer Vaccines , Dendritic Cells/cytology , Neoplasms/metabolism , Neoplasms/therapy , RNA/metabolism , Ultraviolet Rays , Animals , Antigens, Neoplasm , Apoptosis , Bone Marrow Cells/cytology , Female , Humans , Mice , Mice, Inbred C57BL , Papillomaviridae/metabolism , T-Lymphocytes/metabolism
16.
J Virol ; 81(8): 3769-77, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17267484

ABSTRACT

Mouse mammary tumor virus (MMTV) is a milk-transmitted betaretrovirus that causes mammary tumors in mice. Although mammary epithelial cells are the ultimate targets of MMTV, the virus utilizes components of the host immune system to establish infection. Previous studies indicated that dendritic cells play a role in MMTV infection. Here we show that dendritic cells are the first cells to be infected by MMTV in vivo and that they are capable of producing infectious virus that can be transmitted to other cell types. Moreover, upon contact with the virus, dendritic cells became more mature and migrated in response to the chemokine macrophage inflammatory protein 3beta. Finally, we demonstrate that targeted ablation of dendritic cells in vivo dramatically attenuated MMTV infection. These data indicate that MMTV infection of dendritic cells is critical to initial propagation of the virus in vivo.


Subject(s)
Dendritic Cells/virology , Mammary Tumor Virus, Mouse/physiology , Retroviridae Infections/virology , Tumor Virus Infections/virology , Animals , Cell Movement , Dendritic Cells/physiology , Lymph Nodes/virology , Macrophage Inflammatory Proteins/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Transgenic
17.
Cancer Biol Ther ; 5(7): 867-74, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16861891

ABSTRACT

We tested whether tumor cells were killed by replication-incompetent recombinant herpes simplex virus (HSV) d120 lacking immediate early gene ICP4 and whether HSVd120-killed tumor cells could be used directly for tumor vaccination. Vaccine efficacy was tested in TC-1, a murine adenocarcinoma transformed with HPV16 E6 and E7, and ID8-Vegf, a murine epithelial ovarian cancer model. HSVd120 killed tumor cells by apoptosis. Tumor cells infected by HSVd120 were engulfed more avidly by immature DCs and induced DC maturation more efficiently than tumor cells killed by ultraviolet B (UVB) radiation. HSVd120 infection induced stronger upregulation of GRP94 than UVB in cells undergoing apoptosis. Immunization of mice with HSVd120-killed cells elicited stronger antitumor T cell response, including tumor reactive interferon-gamma secreting and cytotoxic T cells, and resulted in significantly stronger delay in tumor growth than immunization with UVB-killed tumor cells. Thus, the use of replication-incompetent HSV strains lacking ICP4 offers possible advantages in the preparation of whole tumor cell antigen for direct tumor vaccination.


Subject(s)
Adenocarcinoma/drug therapy , Antigens, Neoplasm/metabolism , Cancer Vaccines/immunology , Membrane Glycoproteins/metabolism , Ovarian Neoplasms/drug therapy , Simplexvirus/genetics , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Disease Models, Animal , Female , Immediate-Early Proteins/genetics , Membrane Glycoproteins/immunology , Mice , T-Lymphocytes/immunology , Up-Regulation , Vaccination/methods , Virus Replication/genetics
18.
Hum Gene Ther ; 16(6): 765-78, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15960607

ABSTRACT

In the present study, we investigated the ability of replication-restricted herpes simplex virus (HSV) 1716 lacking ICP34.5 to infect endothelium and disrupt tumor vasculature. HSV-1716 efficiently infected and killed mouse endothelial cell lines H5V and MS1 cells, as well as human umbilical vein endothelial cells in vitro. Capillary tube formation by endothelial cells was inhibited by HSV-1716 in vitro and in vivo. Following intratumoral administration of oncolytic HSV-1716, HSV-glycoproteins could be detected in CD31-positive tumor vascular endothelium by immunostaining. Viral DNA was recovered from highly purified microdissected tumor vascular endothelium. Furthermore, endothelium of tumors treated with HSV-1716 exhibited expression of tissue factor, a marker of endothelial damage. Importantly, HSV antigen and DNA were also detected in endothelium distant from foci of active tumor infection. After intravascular inoculation of HSV-1716, viral glycoproteins were detected in association to tumor endothelium, but not vascular endothelium of different organs. Purified tumor endothelial cells showed high proliferative capability and were susceptible to HSV-1716 infection and killing ex vivo while endothelium from normal organs was not. We conclude that oncolytic HSV-1716 exerts direct antiangiogenic effects, which may contribute to the overall therapeutic efficacy of the virus.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carcinoma/therapy , Genetic Vectors/pharmacology , Ovarian Neoplasms/therapy , Simplexvirus/genetics , Angiogenesis Inhibitors/genetics , Animals , Carcinoma/genetics , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Mice , Ovarian Neoplasms/genetics , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/genetics
19.
Mol Ther ; 12(5): 789-802, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15925544

ABSTRACT

Cooperation between oncolytic herpes simplex virus (HSV) and host effector immune mechanisms has been previously described. In the present study, we investigated the mechanism underlying such cooperation in a murine syngeneic model of ovarian carcinoma. Therapeutic administration of HSV-1716, a replication-restricted mutant, resulted in significant reduction of tumor growth and a significant survival advantage. Intratumoral injection of HSV-1716 induced expression of IFN-gamma, MIG, and IP-10 in the tumor. This was accompanied by a significant increase in the number of tumor-associated NK and CD8+ T cells expressing CXCR3 and CD25. Ascites from HSV-1716-treated animals efficiently induced in vitro migration of NK and CD8+ T cells, which was dependent on the presence of MIG and IP-10. Murine monocytes and dendritic cells (DCs) were responsible for the production of MIG and IP-10 upon HSV-1716 infection. In monocytes, this was partially abrogated by neutralizing antibodies against IFN-alpha and -beta, thus indicating a role for type-1 IFNs in the reported effect. Human ovarian carcinomas showed high numbers of monocytes and DCs. Upon HSV-1716 infection, human monocyte-derived DCs produced large amounts of IFN-gamma and upregulated MIG and IP-10 expression. These results indicate that HSV-1716 induces an inflammatory response that may facilitate antitumor immune response upon oncolytic therapy.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Chemokines/metabolism , Genetic Therapy , Ovarian Neoplasms/immunology , Simplexvirus/genetics , Animals , Chemotaxis, Leukocyte , Female , Humans , Interferons/metabolism , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/therapy , Receptors, Chemokine , Up-Regulation
20.
Cancer Res ; 65(3): 925-32, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15705892

ABSTRACT

Epithelial ovarian cancer is the most frequent cause of gynecologic malignancy-related mortality in women. To identify genes up-regulated in ovarian cancer, PCR-select cDNA subtraction was done and Drosophila Eyes Absent Homologue 2 (EYA2) was isolated as a promising candidate. The transcriptional coactivator eya controls essential cellular functions during organogenesis of Drosophila. EYA2 mRNA was found to be up-regulated in ovarian cancer by real-time reverse transcription-PCR, whereas its protein product was detected in 93.6% of ovarian cancer specimens by immunohistochemistry (n = 140). EYA2 was amplified in 14.8% of ovarian carcinomas, as detected by array-based comparative genomic hybridization (n = 88). Most importantly, EYA2 overexpression was significantly associated with short overall survival in advanced ovarian cancer (n = 99, P = 0.0361). EYA2 was found to function as transcriptional activator in ovarian cancer cells by Gal4 assay and to promote tumor growth in vivo in xenograft models. Therefore, this study suggests an important role of EYA2 in ovarian cancer and its potential application as a therapeutic target.


Subject(s)
Ovarian Neoplasms/genetics , Trans-Activators/genetics , Animals , Cell Growth Processes/genetics , DNA, Neoplasm/genetics , Disease Progression , Female , Gene Amplification , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred BALB C , Nuclear Proteins , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prognosis , Protein Tyrosine Phosphatases , Trans-Activators/biosynthesis , Transcriptional Activation/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...