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










Publication year range
1.
Nat Commun ; 14(1): 465, 2023 01 28.
Article in English | MEDLINE | ID: mdl-36709325

ABSTRACT

Oncogenic KRAS expression generates a metabolic dependency on aerobic glycolysis, known as the Warburg effect. We report an effect of increased glycolytic flux that feeds into glycosphingolipid biosynthesis and is directly linked to KRAS oncogenic function. High resolution imaging and genetic approaches show that a defined subset of outer leaflet glycosphingolipids, including GM3 and SM4, is required to maintain KRAS plasma membrane localization, with GM3 engaging in cross-bilayer coupling to maintain inner leaflet phosphatidylserine content. Thus, glycolysis is critical for KRAS plasma membrane localization and nanoscale spatial organization. Reciprocally oncogenic KRAS selectively upregulates cellular content of these same glycosphingolipids, whose depletion in turn abrogates KRAS oncogenesis in pancreatic cancer models. Our findings expand the role of the Warburg effect beyond ATP generation and biomass building to high-level regulation of KRAS function. The positive feedforward loop between oncogenic KRAS signaling and glycosphingolipid synthesis represents a vulnerability with therapeutic potential.


Subject(s)
Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Cell Membrane/metabolism , Signal Transduction , Glycolysis , Glycosphingolipids/metabolism
2.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Article in English | MEDLINE | ID: mdl-34903667

ABSTRACT

KRAS is mutated in 90% of human pancreatic ductal adenocarcinomas (PDACs). To function, KRAS must localize to the plasma membrane (PM) via a C-terminal membrane anchor that specifically engages phosphatidylserine (PtdSer). This anchor-binding specificity renders KRAS-PM localization and signaling capacity critically dependent on PM PtdSer content. We now show that the PtdSer lipid transport proteins, ORP5 and ORP8, which are essential for maintaining PM PtdSer levels and hence KRAS PM localization, are required for KRAS oncogenesis. Knockdown of either protein, separately or simultaneously, abrogated growth of KRAS-mutant but not KRAS-wild-type pancreatic cancer cell xenografts. ORP5 or ORP8 knockout also abrogated tumor growth in an immune-competent orthotopic pancreatic cancer mouse model. Analysis of human datasets revealed that all components of this PtdSer transport mechanism, including the PM-localized EFR3A-PI4KIIIα complex that generates phosphatidylinositol-4-phosphate (PI4P), and endoplasmic reticulum (ER)-localized SAC1 phosphatase that hydrolyzes counter transported PI4P, are significantly up-regulated in pancreatic tumors compared to normal tissue. Taken together, these results support targeting PI4KIIIα in KRAS-mutant cancers to deplete the PM-to-ER PI4P gradient, reducing PM PtdSer content. We therefore repurposed the US Food and Drug Administration-approved hepatitis C antiviral agent, simeprevir, as a PI4KIIIα inhibitor In a PDAC setting. Simeprevir potently mislocalized KRAS from the PM, reduced the clonogenic potential of pancreatic cancer cell lines in vitro, and abrogated the growth of KRAS-dependent tumors in vivo with enhanced efficacy when combined with MAPK and PI3K inhibitors. We conclude that the cellular ER-to-PM PtdSer transport mechanism is essential for KRAS PM localization and oncogenesis and is accessible to therapeutic intervention.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Phosphatidylserines/metabolism , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Receptors, Steroid/metabolism , 1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , 1-Phosphatidylinositol 4-Kinase/genetics , 1-Phosphatidylinositol 4-Kinase/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biological Transport/drug effects , Cell Line, Tumor , Drug Delivery Systems , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Protease Inhibitors/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Steroid/genetics , Simeprevir/pharmacology , Xenograft Model Antitumor Assays
3.
Eur J Med Chem ; 217: 113381, 2021 May 05.
Article in English | MEDLINE | ID: mdl-33756124

ABSTRACT

KRAS plays an essential role in regulating cell proliferation, differentiation, migration and survival. Mutated KRAS is a major driver of malignant transformation in multiple human cancers. We showed previously that fendiline (6) is an effective inhibitor of KRAS plasma membrane (PM) localization and function. In this study, we designed, synthesized and evaluated a series of new fendiline analogs to optimize its drug properties. Systemic structure-activity relationship studies by scaffold repurposing led to the discovery of several more active KRAS PM localization inhibitors such as compounds 12f (NY0244), 12h (NY0331) and 22 (NY0335) which exhibit nanomolar potencies. These compounds inhibited oncogenic KRAS-driven cancer cell proliferation at single-digit micromolar concentrations in vitro. In vivo studies in a xenograft model of pancreatic cancer revealed that 12h and 22 suppressed oncogenic KRAS-expressing MiaPaCa-2 tumor growth at a low dose range of 1-5 mg/kg with no vasodilatory effects, indicating their potential as chemical probes and anticancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane/drug effects , Enzyme Inhibitors/pharmacology , Fendiline/pharmacology , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Female , Fendiline/analogs & derivatives , Fendiline/chemistry , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Structure-Activity Relationship
4.
Mol Cell Biol ; 38(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29158292

ABSTRACT

K-Ras must localize to the plasma membrane (PM) for biological activity. We show here that multiple acid sphingomyelinase (ASM) inhibitors, including tricyclic antidepressants, mislocalized phosphatidylserine (PtdSer) and K-RasG12V from the PM, resulting in abrogation of K-RasG12V signaling and potent, selective growth inhibition of mutant K-Ras-transformed cancer cells. Concordantly, in nude mice, the ASM inhibitor fendiline decreased the rate of growth of oncogenic K-Ras-expressing MiaPaCa-2 tumors but had no effect on the growth of the wild-type K-Ras-expressing BxPC-3 tumors. ASM inhibitors also inhibited activated LET-60 (a K-Ras ortholog) signaling in Caenorhabditis elegans, as evidenced by suppression of the induced multivulva phenotype. Using RNA interference against C. elegans genes encoding other enzymes in the sphingomyelin (SM) biosynthetic pathway, we identified 14 enzymes whose knockdown strongly or moderately suppressed the LET-60 multivulva phenotype. In mammalian cells, pharmacological agents that target these enzymes all depleted PtdSer from the PM and caused K-RasG12V mislocalization. These effects correlated with changes in SM levels or subcellular distribution. Selected compounds, including sphingosine kinase inhibitors, potently inhibited the proliferation of oncogenic K-Ras-expressing pancreatic cancer cells. In conclusion, these results show that normal SM metabolism is critical for K-Ras function, which may present therapeutic options for the treatment of K-Ras-driven cancers.


Subject(s)
Sphingolipids/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Animals , Caenorhabditis elegans , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Humans , Mice , Mice, Nude , Signal Transduction , Sphingomyelins/genetics , Sphingomyelins/metabolism , ras Proteins/metabolism
5.
Biochem Biophys Res Commun ; 483(1): 590-595, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28011265

ABSTRACT

Roquin is an E3 ligase that regulates mRNA stability. Mice with a mutation in the Rc3h1 gene and Roquin protein, referred to as Roquinsan/san or sanroque mice, develop broad-spectrum chronic inflammatory conditions and autoimmune pathologies. Our laboratory recently reported that sanroque mice also develop extensive inflammation that is localized in the small intestine but is rare in the colon. Here, we demonstrate that small intestinal intraepithelial lymphocytes (IELs) are present in the epithelium of sanroque mice but that cell recoverability is low using standard extraction techniques even though lamina propria lymphocytes (LPLs) can be recovered in normal numbers. In studies aimed at characterizing T cell costimulatory markers and activation molecules on LPLs in sanroque mice, we identified Ly6C and 4-1BB (CD137) as being expressed at elevated levels on sanroque small intestinal LPLs, and we show that both of those subsets, in conjunction with cells expressing the KLRG1 T cell activation molecule, are sources of IL-17A, IFN-γ, and TNFα. TNFα was primarily produced by 4-1BB+, KLRG1-cells, but was also made by some 4-1BB-, KLRG1-cells, and 4-1BB-, KLRG1+ cells. These findings collectively suggest that the small intestinal inflammatory response in sanroque mice is driven, at least in part, by LPL activation through Ly6C and 4-1BB signaling, and they provide further evidence in support of using the sanroque mouse as an animal model of chronic small intestinal inflammation.


Subject(s)
Antigens, Ly/physiology , Lymphocytes/metabolism , Mucous Membrane/metabolism , Receptors, Immunologic/physiology , Tumor Necrosis Factor Receptor Superfamily, Member 9/physiology , Animals , Crohn Disease/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Heterozygote , Inflammation , Interferon-gamma/metabolism , Interleukin-17/metabolism , Intestine, Small/metabolism , Lectins, C-Type , Mice , Tumor Necrosis Factor-alpha/metabolism
6.
PLoS One ; 11(1): e0146111, 2016.
Article in English | MEDLINE | ID: mdl-26771831

ABSTRACT

The thyroid stimulating hormone beta-subunit (TSHß) with TSHα form a glycoprotein hormone that is produced by the anterior pituitary in the hypothalamus-pituitary-thyroid (HPT) axis. Although TSHß has been known for many years to be made by cells of the immune system, the role of immune system TSH has remained unclear. Recent studies demonstrated that cells of the immune system produce a novel splice variant isoform of TSHß (TSHßv), but little if any native TSHß. Here, we show that within three days of systemic infection of mice with Listeria monocytogenes, splenic leukocytes synthesized elevated levels of TSHßv. This was accompanied by an influx of CD14+, Ly6C+, Ly6G+ cells into the thyroid of infected mice, and increased levels of intrathyroidal TSHßv gene expression. Adoptive transfer of carboxyfluorescein succinimidyl ester (CFSE)-labeled splenic leukocytes from infected mice into non-infected mice migrated into the thyroid as early as forty-eight hours post-cell transfer, whereas CFSE-labeled cells from non-infected mice failed to traffic to the thyroid. These findings demonstrate for the first time that during bacterial infection peripheral leukocytes produce elevated levels of TSHßv, and that spleen cells traffic to the thyroid where they produce TSHßv intrathyroidally.


Subject(s)
Leukocytes/cytology , Listeria monocytogenes , Listeriosis/metabolism , Protein Isoforms/metabolism , Spleen/cytology , Thyroid Gland/metabolism , Thyrotropin, beta Subunit/metabolism , Animals , Female , Mice , Mice, Inbred C57BL
7.
Gene ; 549(1): 134-40, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25062971

ABSTRACT

Roquin-1, a RING finger E3 ubiquitin ligase, functions as a modulator of inflammation; however, nothing is known about how Rc3h1 expression is regulated. Here, we describe an opposing relationship between Roquin-1 and the IL-17 proinflammatory cytokine by demonstrating that enforced expression of Rc3h1 restricts Il17a expression, and that exposure of T cells to IL-10, a cytokine with immunosuppressive activity, increases Rc3h1 expression. Luciferase reporter assays conducted using eight transcription factor plasmids (STAT1, STAT3, STAT5, GATA2, c-Rel, IKZF1, IKZF2, and IKZF3) demonstrated that STAT1, STAT3, GATA2, and c-Rel increased Rc3h1 promoter activity, whereas IKZF2 decreased activity. Gene expression of those five transcription factors increased in T cells exposed to IL-10. Transcription factor-specific siRNAs suppressed the IL-10 effect on Rc3h1 transcription. These findings identify a role for IL-10 in regulating Rc3h1 transcription, and they have implications for understanding how Roquin-1 controls the immune response.


Subject(s)
Interleukin-10/metabolism , T-Lymphocytes/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Interleukin-17/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics
8.
Sci Rep ; 4: 4920, 2014 May 12.
Article in English | MEDLINE | ID: mdl-24815331

ABSTRACT

Roquin, an E3 ligase, is involved in curtailing autoimmune pathology as seen from studies using mice with mutated (Rc3h1(san/san)) or disrupted (Rc3h1(gt/gt)) Rc3h1 gene. The extent to which intestinal immunopathology is caused by insufficient Roquin expression in the immune system, or by Roquin impairment in non-hematopoietic cells, has not been determined. Using bone marrow cells from Rc3h1(gt/gt) mice transferred into irradiated normal mice (Rc3h1(gt/gt) → NL chimeras), we show that inflammation developed in the small intestine, kidney, lung, liver, and spleen. Proinflammatory cytokine levels were elevated in lamina propria lymphocytes (LPLs). Inflammation in the liver was accompanied by areas of hepatocyte apoptosis. Lung inflammation consisted of an influx of both T cells and B cells. Small intestinal LPLs had increased numbers of CD44(hi), CD62L(lo), KLRG1(+), ICOS(+) short-lived effector cells, indicating an influx of activated T cells. Following oral infection with L. monocytogenes, Rc3h1(gt/gt) → NL chimeras had more liver pathology and greater numbers of bacteria in the Peyer's patches than NL → NL chimeras. These findings demonstrate that small intestinal inflammation in Rc3h1(san/san) and Rc3h1(gt/gt) mice is due to a failure of Roquin expression in the immune system and not to insufficient systemic Roquin expression.


Subject(s)
Enteritis/blood , Enteritis/genetics , Gene Expression , Hematopoiesis/genetics , Ubiquitin-Protein Ligases/genetics , Animals , Antigens, Surface/metabolism , Caspase 3 , Cytokines/metabolism , Disease Models, Animal , Enteritis/immunology , Enteritis/metabolism , Enteritis/pathology , Immunologic Memory , Immunophenotyping , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Intestine, Small/metabolism , Intestine, Small/pathology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Knockout , Peyer's Patches/immunology , Peyer's Patches/metabolism , Ubiquitin-Protein Ligases/deficiency
9.
PLoS One ; 8(2): e56436, 2013.
Article in English | MEDLINE | ID: mdl-23451046

ABSTRACT

Roquin, an E3 ubiquitin ligase that localizes to cytosolic RNA granules, is involved in regulating mRNA stability and translation. Mice that have a M199R mutation in the Roquin protein (referred to as sanroque or Roquin(san/san) mice) develop autoimmune pathologies, although the extent to which these occur in the intestinal mucosa has not been determined. Here, we demonstrate that Roquin(san/san) mice reproducibly develop intestinal inflammation in the small intestine but not the colon. Similarly, mice generated in our laboratory in which the Roquin gene was disrupted by insertion of a gene trap cassette (Roquin(gt/gt) mice) had small intestinal inflammation that mimicked that of Roquin(san/san) mice. MLN cells in Roquin(san/san) mice consisted of activated proliferating T cells, and had increased numbers of CD44(hi) CD62L(lo) KLRG1(+) short-lived effector cells. Proportionally more small intestinal intraepithelial lymphocytes in Roquin(san/san) mice expressed the ICOS T cell activation marker. Of particular interest, small intestinal lamina propria lymphocytes in Roquin(san/san) mice consisted of a high proportion of Gr-1(+) T cells that included IL-17A(+) cells and CD8(+) IFN-γ(+) cells. Extensive cytokine dysregulation resulting in both over-expression and under-expression of chemotactic cytokines occurred in the ileum of Roquin(san/san) mice, the region most prone to the development of inflammation. These findings demonstrate that chronic inflammation ensues in the intestine following Roquin alteration either as a consequence of protein mutation or gene disruption, and they have implications for understanding how small intestinal inflammation is perpetuated in Crohn's disease (CD). Due to the paucity of animal models of CD-like pathophysiology in the small intestine, and because the primary gene/protein defects of the Roquin animal systems used here are well-defined, it will be possible to further elucidate the underlying genetic and molecular mechanisms that drive the disease process.


Subject(s)
Inflammation/immunology , Inflammation/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Animals , Cytokines/immunology , Female , Flow Cytometry , Liver/immunology , Liver/metabolism , Male , Mice , Mice, Mutant Strains , Ubiquitin-Protein Ligases/metabolism
10.
J Immunol ; 187(11): 5834-41, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-22043014

ABSTRACT

IL-10(-/-) mice, an animal model of Th1-mediated inflammatory bowel disease, were screened for the expression of 600 microRNAs (miRNAs) using colonic tissues and PBLs from animals having either mild inflammation or severe intestinal inflammation. The development of colonic inflammation in IL-10(-/-) mice was accompanied by upregulation in the expression of 10 miRNAs (miR-19a, miR-21, miR-31, miR-101, miR-223, miR-326, miR-142-3p, miR-142-5p, miR-146a, and miR-155). Notably, the expression of all of these miRNAs plus miR-375 was elevated in PBLs of IL-10(-/-) mice at a time when colonic inflammation was minimal, suggesting that changes in specific miRNAs in circulating leukocytes may be harbingers of ensuing colonic pathology. In vitro exposure of colonic intraepithelial lymphocytes to IL-10 resulted in downregulation of miR-19a, miR-21, miR-31, miR-101, miR-223, and miR-155. Interestingly, unlike IL-10(-/-) mice, changes in miRNAs in PBL of dextran sulfate sodium-treated mice were minimal but selectively elevated in the colon after pathology was severe. We further show that miR-223 is a negative regulator of the Roquin ubiquitin ligase, Roquin curtails IL-17A synthesis, and the 3' untranslated region of Roquin is a target for miR-223, thus defining a molecular pathway by which IL-10 modulates IL-17-mediated inflammation. To identify additional miRNAs that may be involved in the regulation of Roquin, transcriptome analysis was done using cDNAs from HeLa cells transfected with 90 miRNA mimics. Twenty-six miRNAs were identified as potential negative regulators of Roquin, thus demonstrating functional complexity in gene expression regulation by miRNAs.


Subject(s)
Colon/metabolism , Gene Expression Regulation/genetics , Inflammatory Bowel Diseases/genetics , Leukocytes/metabolism , MicroRNAs/analysis , Animals , Colon/immunology , Colon/pathology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Profiling , Gene Expression Regulation/immunology , HeLa Cells , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin-10/deficiency , Interleukin-10/immunology , Leukocytes/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , MicroRNAs/biosynthesis , MicroRNAs/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Up-Regulation
11.
J Periodontol ; 81(12): 1850-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20843233

ABSTRACT

BACKGROUND: A major cause of chronic inflammatory periodontal disease is Porphyromonas gingivalis, a non-motile, Gram-negative, rod-shaped, anaerobic bacterium. Within gingival tissue, both macrophages and fibroblasts participate in the immune response to foreign entities by releasing cytokines and expressing molecules to recruit and activate lymphocytes. However, the contribution of gingival macrophages and fibroblasts to the immune response to P. gingivalis infection is not fully known. METHODS: The AMJ2-C8 cell line (AM cells), a mouse alveolar macrophage cell line, and ESK-1 cells, a mouse gingival fibroblast cell line made in our laboratory, were treated with lipopolysaccharide (LPS) from either P. gingivalis or Escherichia coli. The expression of immune response molecules was quantified by real-time polymerase chain reaction and enzyme-linked immunoassay. RESULTS: AM and ESK-1 cells responded differently to P. gingivalis and E. coli LPS stimulation. The ESK-1 gingival fibroblast cell line was more responsive to E. coli LPS stimulation as seen by elevated levels of interleukin (IL)-6, inducible nitric oxide, and monocyte chemotactic protein-1 expression relative to stimulation by P. gingivalis LPS. Conversely, the AM macrophage cell line was more responsive to P. gingivalis LPS stimulation, particularly for interleukin IL-1ß, IL-6, and monocyte chemotactic protein-1, relative to stimulation by E. coli LPS. CONCLUSION: These findings demonstrate that E. coli LPS induces a stronger cytokine and chemokine response in gingival fibroblasts, whereas P. gingivalis LPS induces a stronger response in macrophages.


Subject(s)
Cytokines/immunology , Escherichia coli/immunology , Fibroblasts/immunology , Gingiva/immunology , Lipopolysaccharides/immunology , Macrophages, Alveolar/immunology , Porphyromonas gingivalis/immunology , Animals , Cell Line, Transformed , Chemokine CCL2/analysis , Chemokine CCL2/immunology , Chemokine CCL3/analysis , Chemokine CCL3/immunology , Chemokine CCL4/analysis , Chemokine CCL4/immunology , Cytokines/analysis , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/microbiology , Gingiva/cytology , Gingiva/microbiology , Interleukin-1beta/analysis , Interleukin-1beta/immunology , Interleukin-23/analysis , Interleukin-23/immunology , Interleukin-6/analysis , Macrophages, Alveolar/microbiology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Nitric Oxide Synthase Type II/analysis , Nitric Oxide Synthase Type II/immunology , Reverse Transcriptase Polymerase Chain Reaction
12.
Int J Exp Pathol ; 91(3): 276-80, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20353422

ABSTRACT

Soluble gp130 (sgp130) has been shown to suppress the inflammatory response of autoimmune pathologies; however, its effects on virus infection are not known. Here, we report that intraperitoneal treatment of mice with sgp130-Fc fusion protein at the time of oral reovirus serotype 3 infection resulted in altered morphopathological changes that were evident by less shortening of intestinal villi length and crypt depth after infection. That the effect mediated by sgp130 treatment was due to an increase in intestinal crypt cell proliferation was demonstrated by an increase in the number of crypt mitotic figures. This was further confirmed by increased immunoreactivity to the Cdc47 proliferation-associated antigen in crypts of sgp130-treated virus-infected mice compared to infected non-treated mice. These findings suggest that sgp130 may have a beneficial effect during intestinal virus infection by disrupting interleukin-6 trans-signalling, thereby reducing the local inflammatory response.


Subject(s)
Cytokine Receptor gp130/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Reoviridae Infections/pathology , Animals , Cytokine Receptor gp130/immunology , Female , Hyperplasia , Inflammation/immunology , Inflammation/virology , Intestinal Mucosa/metabolism , Mammalian orthoreovirus 3/immunology , Mice , Mice, Inbred C57BL , Reoviridae Infections/immunology , Reoviridae Infections/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology
13.
J Leukoc Biol ; 87(2): 301-8, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19889730

ABSTRACT

In the absence of IL-10, colonic inflammation ensues, which is characterized by high levels of IL-17. Here, we demonstrate a direct correlation between ICOS expression and IL-17 production in cIELs. IL-10(-/-) mice had increased numbers of cIELs and greater colon weight. Although the CD69 early activation antigen was expressed on cIELs from normal and IL-10(-/-) mice, ICOS was expressed only on cIELs from IL-10(-/-) mice. IL-17-producing cells in IL-10(-/-) mice consisted of CD4(+) and CD8(+) cIELs; however, CD4(+) cells were the predominant IL-17-producing cell population. Culture of cIELs from IL-10(-/-) mice with IL-23 resulted in an increase in ICOS and IL-17 expression, whereas IL-10 suppressed expression of ICOS and IL-17. This occurred in primary cultures and recall stimulation experiments. The ICOS ligand B7RP-1 was up-regulated on colonic epithelial cells and on a population of large granular leukocytes during inflammation. Culture of cIELs with B7RP-1(+) DCs enhanced IL-17A production from normal cIELs but failed to do so using cIELs from ICOS(-/-) mice. In vivo treatment of IL-10(-/-) mice with antibody to ICOS resulted in a significant reduction in colonic pathology. These findings implicate ICOS as an activational signal of Th17 cells during chronic intestinal inflammation, and they suggest that under some conditions, control of ICOS expression may help to suppress chronic intestinal inflammation.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Colon/immunology , Interleukin-10/immunology , Interleukin-17/immunology , Intestinal Mucosa/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/metabolism , B7-1 Antigen/genetics , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Colon/metabolism , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Inflammation/immunology , Inflammation/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-17/biosynthesis , Interleukin-17/genetics , Intestinal Mucosa/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout
14.
J Cell Biochem ; 105(1): 271-6, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18523983

ABSTRACT

The small intestine has been shown to be an extra-pituitary site of thyroid stimulating hormone (TSH) production, and previous in vivo studies have shown that TSH synthesis localizes within areas of enteric virus infection within the small intestine; however, the cellular source of intestinal TSH has not been adequately determined. In the present study, we have used the murine MODE-K small intestinal epithelial cell line to demonstrate both at the transcriptional level and as a secreted hormone, as measured in a TSHbeta-specific enzyme-linked assay, that epithelial cells in fact respond to infection with reovirus serotype 3 Dearing strain by upregulating TSH synthesis. Moreover, sequence analysis of a PCR-amplified TSHbeta product from MODE-K cells revealed homology to mouse pituitary TSHbeta. These findings have direct functional implications for understanding a TSH immune-endocrine circuit in the small intestine.


Subject(s)
Epithelial Cells/metabolism , Intestines/cytology , Mammalian orthoreovirus 3/physiology , Thyrotropin, beta Subunit/biosynthesis , Animals , Cell Line , Mice , Sequence Analysis , Thyrotropin, beta Subunit/genetics , Thyrotropin, beta Subunit/metabolism , Up-Regulation
15.
Int Immunol ; 20(1): 141-54, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18046045

ABSTRACT

IL-10-deficient mice develop enterocolitis due to a failure of cytokine regulation; however, the full scope of that response remains poorly defined. Using multiplex analysis to quantify the activity of 23 regulatory and effector cytokines produced by colonic leukocytes, we demonstrate a vast dysregulation process of 18 cytokines in IL-10-/- mice from 7 to 27 weeks of age. Of those, IL-12p40, IL-6, granulocyte macrophage colony-stimulating factor, IFN-gamma, IL-13 and monocyte chemoattractant protein-1 (MCP-1) had the highest single correlations with pathology (r = 0.7766-0.7016). Importantly, there were strong associations (r = 0.7071-0.9074) between those cytokines and as many as 10 additional cytokines, indicating a high degree of cytokine complexity as disease progressed. IL-17 was notable in that it was produced at high levels by colonic leukocytes from IL-10-/- mice with pathology ranging from mild to severe, though it was not produced by healthy IL-10-/- mice lacking pathology. Tumor necrosis factor alpha (TNFalpha) by itself displayed only a modest association with pathology (r = 0.6340), ranking sixth lowest, though it cross-correlated strongly with the synthesis of 12 other cytokines, implying that the destructive effects associated with TNFalpha may be due to interactions of multiple cytokine activities. IL-23 expression did not correlate with pathology, possibly suggesting that IL-23 is involved in the initiation but not the perpetuation of inflammation. Four cytokines (IL-2, IL-3, IL-4 and IL-5) remained negative in IL-10-/- mice, demonstrating that cytokine dysregulation was not universal. These findings emphasize the need to better understand cytokine networks in chronic inflammation and they provide a rationale for combining immunotherapies in the treatment of intestinal inflammation.


Subject(s)
Colitis/immunology , Colitis/physiopathology , Colon/pathology , Cytokines/metabolism , Epithelial Cells/pathology , Interleukin-10/deficiency , Leukocytes/pathology , Animals , Colitis/pathology , Colon/cytology , Colon/immunology , Cytokines/genetics , Cytokines/immunology , Epithelial Cells/cytology , Epithelial Cells/immunology , Female , Interleukin-10/genetics , Leukocytes/cytology , Leukocytes/immunology , Male , Mice , Mice, Inbred BALB C
16.
J Leukoc Biol ; 82(5): 1166-73, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17702824

ABSTRACT

This study has examined the stimulatory and costimulatory effects of IL-18 on two subsets of murine small intestinal intraepithelial lymphocytes (IELs) defined by the expression of the CD43 S7 glycoform. Data from gene array studies and real-time PCR indicated that S7(+) IELs had significantly higher levels of gene expression for the IL-18 receptor and the IL-18R accessory protein than S7(-) IELs. IL-18 costimulation of IELs in conjunction with CD3-induced activation resulted in significantly greater proliferation than CD3 stimulation alone. In CFSE dilution experiments, IL-18 costimulation favored the S7(+) IEL population. IL-18 costimulation did not affect apoptosis of either S7(-) or S7(+) IELs compared with CD3 stimulation alone. Although IL-18 costimulation did not alter the total number of IFN-gamma-producing cells relative to CD3 stimulation alone, twice as many S7(+) IELs were IFN-gamma -secreting cells than S7(-) IELs in both CD3-stimulated and IL-18-costimulated cultures. Notably, direct IL-18 stimulation in the absence of CD3 activation induced an IFN-gamma response that was predominantly directed to the S7(+) population, indicating that IL-18 is itself an IFN-gamma activational signal for intestinal T cells. In contrast, direct IL-18 stimulation of IELs did not generate TNF-alpha-producing cells, indicating a differential response in the activation of proinflammatory cytokines following IL-18 exposure. These findings point to distinctly different activational effects of IL-18 on IELs, both with regard to the type of functional responses elicited and with respect to the IEL subsets affected.


Subject(s)
Interleukin-18/pharmacology , Intestinal Mucosa/immunology , Intestine, Small/immunology , Leukosialin/metabolism , T-Lymphocyte Subsets/immunology , Animals , CD3 Complex/immunology , CD3 Complex/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Death/immunology , Cell Proliferation , Flow Cytometry , Interferon-gamma/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestine, Small/cytology , Intestine, Small/metabolism , Leukosialin/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Receptors, Interleukin-18/metabolism , T-Lymphocyte Subsets/metabolism
17.
Immunol Rev ; 215: 189-201, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17291289

ABSTRACT

The vast majority of peripheral T cells exist as resting lymphocytes until a signal for activation has been received. In response to antigen, this activation involves ligation of the T-cell receptor (TCR) and signal transmission through the CD3 complex, which then initiates a cascade of intracellular events that lead to the expression of genes used in T-cell activation. T-cell activation also requires soluble mediators in the form of cytokines and chemokines that regulate the process in both positive and negative ways, and costimulatory signals received in conjunction with TCR/CD3 signaling are important in the activation of T cells. Unlike T cells in other peripheral immune compartments, small and large intestinal intraepithelial lymphocytes (IELs) bear some but not all properties of activated T cells, suggesting that they constitute a large population of 'partially activated' effector cells. Thus, regulation of the IEL activation process must be held in tight check, yet it must be ready to respond to foreign antigen rapidly and effectively. We discuss how costimulatory molecules may hold the key to controlling IEL activation through a multiphase process beginning with cells that have already entered into the early stage of activation.


Subject(s)
Immunity, Mucosal , Intestinal Mucosa/immunology , Lymphocyte Activation/immunology , Models, Immunological , T-Lymphocytes/immunology , Animals , Epithelial Cells/cytology , Epithelial Cells/immunology , Humans , Intestinal Mucosa/cytology
18.
Anat Rec A Discov Mol Cell Evol Biol ; 288(3): 316-22, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16463380

ABSTRACT

Achondroplasia, the most common short-limbed dwarfism in humans, results from a single nucleotide substitution in the gene for fibroblast growth factor receptor 3 (FGFR3). FGFR3 regulates bone growth in part via the mitogen-activated protein kinase pathway (MAPK). To examine the role of this pathway in chondrocyte differentiation, a transgenic mouse was generated that expresses a constitutively active mutant of MEK1 in chondrocytes and exhibits dwarfing characteristics typical of human achondroplasia, i.e., shortened axial and appendicular skeletons, mid-facial hypoplasia, and dome-shaped cranium. In this study, cephalometrics of the MEK1 mutant skulls were assessed to determine if the MEK1 mice are a good model of achondroplasia. Skull length, arc of the cranial vault, and area, maximum and minimum diameters of the brain case were measured on digitized radiographs of skulls of MEK1 and control mice. Cranial base and nasal bone length and foramen magnum diameter were measured on midsagittal micro-CT sections. Data were normalized by dividing by the cube root of each animal's weight. Transgenic mice exhibited a domed skull, deficient midface, and (relatively) prognathic mandible and had a shorter cranial base and nasal bone than the wild-type. Skull length was significantly less in transgenic mice, but cranial arc was significantly greater. The brain case was larger and more circular and minimum diameter of the brain case was significantly greater in transgenic mice. The foramen magnum was displaced anteriorly but not narrowed. MEK1 mouse cephalometrics confirm these mice as a model for achondroplasia, demonstrating that the MAP kinase signaling pathway is involved in FGF signaling in skeletal development.


Subject(s)
Achondroplasia/pathology , Disease Models, Animal , Mice, Transgenic/abnormalities , Skull/pathology , Achondroplasia/diagnostic imaging , Achondroplasia/genetics , Animals , Cephalometry , Mice , Radiography , Skull/diagnostic imaging
19.
Biochem Biophys Res Commun ; 342(3): 782-90, 2006 Apr 14.
Article in English | MEDLINE | ID: mdl-16500623

ABSTRACT

Murine intestinal intraepithelial lymphocytes (IELs) can be classified according to expression of a CD43 glycoform recognized by the S7 monoclonal antibody. In this study, we examined the response of S7+ and S7- IELs in mice during acute reovirus serotype 3 (Dearing strain) infection, which was confirmed by virus-specific real-time PCR. In vivo proliferation increased significantly for both S7- and S7+ IELs on day 4 post-infection as determined by BrdU incorporation; however, expression of the inducible costimulatory (ICOS) molecule, which peaked on day 7 post-infection, was upregulated on S7+ CD4+ T cells, most of which were CD4+8- IELs. In vitro ICOS stimulation by syngeneic peritoneal macrophages induced IFN-gamma secretion from IELs from day 7 infected mice, and was suppressed by treatment with anti-ICOS mAb. Additionally, IFN-gamma mRNA increased in CD4+ IELs on day 6 post-infection. These findings indicate that S7- and S7+ IELs are differentially mobilized during the immune response to reovirus infection; that the regulated expression of ICOS is associated with S7+ IELs; and that stimulation of IELs through ICOS enhances IFN-gamma synthesis during infection.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/metabolism , Epithelial Cells/metabolism , Intestine, Small/cytology , Leukosialin/metabolism , Reoviridae Infections/metabolism , Up-Regulation/genetics , Animals , Antibodies, Monoclonal , B7-1 Antigen/metabolism , Cell Proliferation , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Flow Cytometry , Inducible T-Cell Co-Stimulator Ligand , Inducible T-Cell Co-Stimulator Protein , Interferon-gamma/biosynthesis , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL
20.
J Oral Maxillofac Surg ; 64(2): 243-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16413896

ABSTRACT

PURPOSE: Surprisingly little is known about the cellular composition of the temporomandibular joint (TMJ) disc, which is a crucial piece of the puzzle in tissue engineering efforts. Toward this end, cell types were identified and quantified regionally in the TMJ disc. MATERIALS AND METHODS: Porcine TMJ discs were examined by histology, electron microscopy, and immunohistochemistry. Histology consisted of hematoxylin and eosin staining to identify regional variation of cell type and cell numbers. Transmission electron microscopy was used to elucidate differences in organelle content and pericellular matrix between TMJ disc cells and chondrocytes from hyaline cartilage. Immunohistochemistry was used to assess the presence of smooth and skeletal muscle character in the TMJ disc. RESULTS: The overall ratio of fibroblasts to chondrocyte-like cells in the TMJ disc was approximately 2.35 to 1, with the highest relative number of chondrocyte-like cells in the intermediate zone. Electron microscopy revealed distinct differences between TMJ disc chondrocyte-like cells and chondrocytes from hyaline cartilage with respect to organelles and the pericellular region. Immunostaining identified smooth muscle in the form of vessels, which were most prominent in the anterior band. Skeletal muscle was not observed. CONCLUSION: The cells of the TMJ disc are distinctly different from cells of hyaline cartilage, and consequently should not be referred to as chondrocytes. TMJ disc cells are comprised of heterogeneously distributed subpopulations, with fibroblasts predominating over fibrochondrocytes.


Subject(s)
Chondrocytes , Fibroblasts , Temporomandibular Joint Disc/cytology , Animals , Cell Count , Chondrocytes/ultrastructure , Female , Fibroblasts/ultrastructure , Hyaline Cartilage/cytology , Microscopy, Electron, Scanning Transmission , Swine , Temporomandibular Joint Disc/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...