Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Gastroenterology ; 160(4): 1240-1255.e3, 2021 03.
Article in English | MEDLINE | ID: mdl-33189700

ABSTRACT

BACKGROUND & AIMS: The down-regulated in adenoma (DRA) protein, encoded by SLC26A3, a key intestinal chloride anion exchanger, has recently been identified as a novel susceptibility gene for inflammatory bowel disease (IBD). However, the mechanisms underlying the increased susceptibility to inflammation induced by the loss of DRA remain elusive. Compromised barrier is a key event in IBD pathogenesis. The current studies were undertaken to elucidate the impact of DRA deficiency on epithelial barrier integrity and to define underlying mechanisms. METHODS: Wild-type and DRA-knockout (KO) mice and crypt-derived colonoids were used as models for intestinal epithelial response. Paracellular permeability was measured by using fluorescein isothiocyanate-dextran flux. Immunoblotting, immunofluorescence, immunohistochemistry, and ribonucleoprotein immunoprecipitation assays were performed. Gut microbiome analysis was conducted to investigate the impact of DRA deficiency on gut microbial communities. RESULTS: DRA-KO mice exhibited an increased colonic paracellular permeability with significantly decreased levels of tight junction/adherens junction proteins, including ZO-1, occludin, and E-cadherin. A similar expression pattern of occludin and E-cadherin was observed in colonoids derived from DRA-KO mice and short hairpin RNA-mediated DRA knockdown in Caco-2 cells. Microbial analysis showed gut dysbiosis in DRA-KO mice. However, cohousing studies showed that dysbiosis played only a partial role in maintaining tight junction protein expression. Furthermore, our results showed increased binding of RNA-binding protein CUGBP1 with occludin and E-cadherin genes in DRA-KO mouse colon, suggesting that posttranscriptional mechanisms play a key role in gut barrier dysfunction. CONCLUSIONS: To our knowledge, our studies demonstrate a novel role of DRA in maintaining the intestinal epithelial barrier function and potential implications of its dysregulation in IBD pathogenesis.


Subject(s)
Antiporters/deficiency , Chloride-Bicarbonate Antiporters/deficiency , Dysbiosis/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Sulfate Transporters/deficiency , Animals , Antiporters/genetics , CELF1 Protein/metabolism , Caco-2 Cells , Cadherins/metabolism , Chloride-Bicarbonate Antiporters/genetics , Disease Models, Animal , Dysbiosis/microbiology , Dysbiosis/pathology , Gene Knockdown Techniques , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Male , Mice , Mice, Knockout , Occludin/metabolism , Permeability , Sulfate Transporters/genetics , Tight Junctions/pathology
2.
Anal Biochem ; 640: 114455, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34788604

ABSTRACT

We recently demonstrated that capturing human monoclonal antibodies (hmAbs) using high affinity anti-human Fc (AHC) antibodies allows reliable characterization of antibody-antigen interactions. Here, we characterized six human Fc specific mouse monoclonal antibodies (mAbs) and compared their binding profiles with three previously characterized goat AHC polyclonal antibodies (pAbs), exhibiting properties of a good capture reagent. All six mouse AHC mAbs specifically bound with high affinity to the Fc region of hIgG1, hIgG2, hIgG4 and to 43 different hIgG variants, containing substitutions and/or mutations in the hinge and/or Fc region, that have been reported to exhibit modified antibody effector function and/or pharmacokinetics. Biacore sensor surfaces individually derivatized with mouse AHC mAbs exhibited >2.5-fold higher hIgG binding capacity compared to the three goat AHC pAb surfaces and reproducibly captured hIgG over 300 capture-regeneration cycles. The results of the capture kinetic analyses performed on 31 antibody-antigen interactions using surfaces derivatized with either of the two highest affinity AHC mAbs (REGN7942 or REGN7943) were in concordance with those performed using goat AHC pAb surfaces. Our data demonstrate that AHC mAbs such as REGN7942 and REGN7943 that have properties superior than the three goat AHC pAbs are highly valuable research reagents, especially to perform capture kinetic analyses of antibody-antigen interactions on optical biosensors.


Subject(s)
Antibodies, Monoclonal
3.
Am J Physiol Cell Physiol ; 317(6): C1205-C1212, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31483700

ABSTRACT

The protozoan parasite Cryptosporidium parvum (CP) causes cryptosporidiosis, a diarrheal disease worldwide. Infection in immunocompetent hosts typically results in acute, self-limiting, or recurrent diarrhea. However, in immunocompromised individuals infection can cause fulminant diarrhea, extraintestinal manifestations, and death. To date, the mechanisms underlying CP-induced diarrheal pathogenesis are poorly understood. Diarrheal diseases most commonly involve increased secretion and/or decreased absorption of fluid and electrolytes. We and others have previously shown impaired chloride absorption in infectious diarrhea due to dysregulation of SLC26A3 [downregulated in adenoma (DRA)], the human intestinal apical membrane Cl-/HCO3- exchanger protein. However, there are no studies on the effects of CP infection on DRA activity. Therefore, we examined the expression and function of DRA in intestinal epithelial cells in response to CP infection in vitro and in vivo. CP infection (0.5 × 106 oocysts/well in 24-well plates, 24 h) of Caco-2 cell monolayers significantly decreased Cl-/HCO3- exchange activity (measured as DIDS-sensitive 125I uptake) as well as DRA mRNA and protein levels. Substantial downregulation of DRA mRNA and protein was also observed following CP infection ex vivo in mouse enteroid-derived monolayers and in vivo in the ileal and jejunal mucosa of C57BL/6 mice for 24 h. However, at 48 h after infection in vivo, the effects on DRA mRNA and protein were attenuated and at 5 days after infection DRA returned to normal levels. Our results suggest that impaired chloride absorption due to downregulation of DRA could be one of the contributing factors to CP-induced acute, self-limiting diarrhea in immunocompetent hosts.


Subject(s)
Antiporters/genetics , Chloride-Bicarbonate Antiporters/genetics , Cryptosporidiosis/genetics , Cryptosporidium parvum/pathogenicity , Gene Expression Regulation/genetics , Intestinal Mucosa/metabolism , Sulfate Transporters/genetics , Animals , Antibodies, Neutralizing/pharmacology , Antiporters/antagonists & inhibitors , Antiporters/metabolism , Caco-2 Cells , Chloride-Bicarbonate Antiporters/antagonists & inhibitors , Chloride-Bicarbonate Antiporters/metabolism , Chlorides/metabolism , Cryptosporidiosis/metabolism , Cryptosporidiosis/parasitology , Cryptosporidium parvum/physiology , Host-Parasite Interactions/genetics , Humans , Ileum/metabolism , Ileum/parasitology , Intestinal Mucosa/parasitology , Ion Transport , Janus Kinases/genetics , Janus Kinases/metabolism , Mice , Mice, Inbred C57BL , Organoids/metabolism , Organoids/parasitology , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Sulfate Transporters/antagonists & inhibitors , Sulfate Transporters/metabolism
4.
Cell Microbiol ; 20(6): e12830, 2018 06.
Article in English | MEDLINE | ID: mdl-29444370

ABSTRACT

Infection with the protozoan parasite Cryptosporidium parvum (CP) causes cryptosporidiosis, a widespread diarrhoeal disease. Impaired intestinal epithelial barrier function and increased permeability are most commonly associated with diarrhoeal diseases caused by enteric infections. However, studies on barrier disruption and underlying mechanisms in cryptosporidiosis are extremely limited. Epithelial tight junctions (TJs) and adherens junctions (AJs) are important in maintaining barrier integrity. Therefore, we examined the effects of CP infection on paracellular permeability and on the expression of the major TJ and AJ proteins utilising in vitro, ex vivo, and in vivo models. CP infection (0.5 × 106  oocysts/well in Transwell inserts, 24 hr) increased paracellular permeability (FITC-dextran flux) in Caco-2 cell monolayers and substantially decreased the protein levels of occludin, claudin 4, and E-cadherin. Claudin 3, zonula occludens-1 (ZO1) and α-catenin were also significantly decreased, whereas claudins 1 and 2 and ß-catenin were not altered. Substantial downregulation of occludin, claudin 4, and E-cadherin was also observed in response to CP infection ex vivo in mouse enteroid-derived monolayers and in vivo in the ileal and jejunal mocosa of C57BL/6 mice. The mRNA levels of these proteins were also significantly decreased in CP-infected mouse ileum and jejunum but were unaltered in Caco-2 cells. Further, bafilomycin-A, an inhibitor of lysosomal proton pump, partially abrogated CP effects on occludin expression in Caco-2 cells, suggesting a potential role of posttranslational mechanisms, such as induction of protein degradation pathways, in mediating the effects of the parasite. Our studies suggest that disruption of barrier function via downregulation of specific key components of TJ and AJ could be a major mechanism underlying CP infection-induced diarrhoea.


Subject(s)
Adherens Junctions/parasitology , Cell Adhesion Molecules/antagonists & inhibitors , Cryptosporidiosis/pathology , Cryptosporidium parvum/growth & development , Down-Regulation , Host-Pathogen Interactions , Tight Junctions/parasitology , Animals , Caco-2 Cells , Disease Models, Animal , Gene Expression Profiling , Humans , Intestinal Mucosa/parasitology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Models, Biological , Permeability
5.
Gastroenterology ; 153(5): 1338-1350.e3, 2017 11.
Article in English | MEDLINE | ID: mdl-28823863

ABSTRACT

BACKGROUND & AIMS: Diarrhea associated with inflammatory bowel diseases has been associated with increased levels of inflammatory cytokines, including tumor necrosis factor (TNF). The intestinal mucosa of patients with inflammatory bowel diseases has reduced expression of solute carrier family 26 member 3 (SLC26A3, also called DRA). We investigated whether TNF directly affects expression of DRA in human intestinal epithelial cells (IECs) and in the intestines of mice, and studied the mechanisms of these effects. METHODS: We performed quantitative reverse transcription polymerase chain reaction, immunofluorescence, and immunoblot analyses in Caco-2, HT-29, and T-84 cells human IECs cultured in 2 or 3 dimensions with or without TNF (50 ng/mL for 6-24 hours). We purified nuclear extracts and quantified nuclear factor-κB (NF-κB) activation and DNA binding. We isolated intestinal crypts from C57BL/6 mice, cultured enteroids, incubated these with TNF (50 ng/mL, 24 hours), and quantified messenger RNAs. DRA-mediated exchange of Cl- for HCO3- was measured by uptake of 125I. Expression of the NF-κB inhibitor α (IkBa) was knocked down in Caco-2 cells with small interfering RNAs. Activation of NF-κB in response to TNF was measured by luciferase reporter assays; binding of the NF-κB subunit p65 in cells was analyzed in chromatin immunoprecipitation assays. DRA promoter activity was measured in a luciferase reporter assay. C57BL/6 mice were injected with TNF (5 µg/mouse for 3-6 hours) or vehicle (control); intestines were collected and analyzed by immunofluorescence, or RNA and protein were collected from the mucosa. RESULTS: Incubation of IECs with TNF reduced expression of DRA. Knockdown of NF-κB inhibitor α in IECs led to nuclear translocation of the NF-κB subunit p65 and reduced levels of DRA messenger RNA and protein. Expression of a transgene encoding p65 or p50 in IECs led to significant reductions in the promoter activity of DRA and its expression. In chromatin immunoprecipitation assays, p65 bound directly to the promoter of DRA, at the regions of -935 to -629 and -375 to -84. Injection of mice with TNF or incubation of crypt-derived enteroids with TNF reduced their expression of DRA messenger RNA and protein. CONCLUSIONS: In human IECs and intestinal tissues from mice, we found TNF to activate NF-κB, which reduced expression of the Cl- / HCO3- exchanger DRA (SLC26A3), via direct binding to the promoter of DRA. This pathway is an important therapeutic target for inflammatory bowel disease-associated diarrhea.


Subject(s)
Antiporters/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Diarrhea/etiology , Epithelial Cells/drug effects , Inflammatory Bowel Diseases/complications , Intestinal Mucosa/drug effects , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Antiporters/genetics , Caco-2 Cells , Chloride-Bicarbonate Antiporters/genetics , Diarrhea/genetics , Diarrhea/metabolism , Dose-Response Relationship, Drug , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , HT29 Cells , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Promoter Regions, Genetic , RNA Interference , Sulfate Transporters , Time Factors , Transfection
6.
Am J Physiol Gastrointest Liver Physiol ; 313(3): G256-G264, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28572085

ABSTRACT

SLC26A3 [downregulated in adenoma (DRA)] plays a key role in mammalian intestinal NaCl absorption, in that it mediates apical membrane Cl-/[Formula: see text] exchange. DRA function and expression are significantly decreased in diarrhea associated with inflammatory bowel disease. DRA is also considered to be a marker of cellular differentiation and is predominantly expressed in differentiated epithelial cells. Caudal-type homeobox protein-2 (CDX2) is known to regulate genes involved in intestinal epithelial differentiation and proliferation. Reduced expression of both DRA and CDX2 in intestinal inflammation prompted us to study whether the DRA gene is directly regulated by CDX2. Our initial studies utilizing CDX2 knockout (CDX2fV/fV;Cre+) mice showed a marked reduction in DRA mRNA and protein levels in proximal and distal colon. In silico analysis of the DRA promoter showed two consensus sites for CDX2 binding. Therefore, we utilized Caco-2 cells as an in vitro model to examine if DRA is a direct target of CDX2 regulation. siRNA-mediated silencing of CDX2 in Caco-2 cells resulted in a marked (~50%) decrease in DRA mRNA and protein levels, whereas ectopic overexpression of CDX2 upregulated DRA expression and also stimulated DRA promoter activity, suggesting transcriptional regulation. Electrophoretic mobility shift and chromatin immunoprecipitation assays demonstrated direct binding of CDX2 to one of the two putative CDX2 binding sites in the DRA promoter (+645/+663). In summary, our studies, for the first time, demonstrate transcriptional regulation of DRA expression by CDX2, implying that reduced expression of DRA in inflammatory bowel disease-associated diarrhea may, in part, be due to downregulation of CDX2 in the inflamed mucosa.NEW & NOTEWORTHY SLC26A3 [downregulated in adenoma (DRA)] mediates intestinal luminal NaCl absorption and is downregulated in inflammatory bowel disease-associated diarrhea. Since both DRA and caudal-type homeobox protein-2 (CDX2) are reduced in intestinal inflammation and the DRA promoter harbors CDX2 binding sites, we examined whether the DRA gene is regulated by CDX2. Our studies, for the first time, demonstrate transcriptional regulation of DRA expression by CDX2 via direct binding to the DRA promoter, suggesting that reduced expression of DRA in inflammatory bowel disease-associated diarrhea could, in part, be attributed to downregulation of CDX2.


Subject(s)
Antiporters/metabolism , CDX2 Transcription Factor/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Animals , Antiporters/genetics , CDX2 Transcription Factor/genetics , Caco-2 Cells , Chloride-Bicarbonate Antiporters/genetics , Gene Expression Regulation/physiology , Humans , Mice , RNA Interference , RNA, Small Interfering , Sulfate Transporters
7.
Nanomedicine ; 13(2): 659-665, 2017 02.
Article in English | MEDLINE | ID: mdl-27553076

ABSTRACT

The gut hormone, glucagon like peptide-1 (GLP-1) exerts anti-inflammatory effects. However, its clinical use is limited by its short half-life. Previously, we have shown that GLP-1 as a nanomedicine (GLP-1 in sterically stabilized phospholipid micelles, GLP-1-SSM) has increased in vivo stability. The current study was aimed at testing the efficacy of this GLP-1 nanomedicine in alleviating colonic inflammation and associated diarrhea in dextran sodium sulfate (DSS) induced mouse colitis model. Our results show that GLP-1-SSM treatment markedly alleviated the colitis phenotype by reducing the expression of pro-inflammatory cytokine IL-1ß, increasing goblet cells and preserving intestinal epithelial architecture in colitis model. Further, GLP-1-SSM alleviated diarrhea (as assessed by luminal fluid) by increasing protein expression of intestinal chloride transporter DRA (down regulated in adenoma). Our results indicate that GLP-1 nanomedicine may act as a novel therapeutic tool in alleviating gut inflammation and associated diarrhea in inflammatory bowel disease (IBD).


Subject(s)
Colitis/drug therapy , Glucagon-Like Peptide 1/administration & dosage , Inflammation , Nanomedicine , Animals , Dextran Sulfate/therapeutic use , Diarrhea/drug therapy , Diarrhea/etiology , Disease Models, Animal , Glucagon-Like Peptide 1/therapeutic use , Mice
8.
Am J Physiol Gastrointest Liver Physiol ; 311(5): G817-G826, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27634011

ABSTRACT

Impaired absorption of electrolytes is a hallmark of diarrhea associated with inflammation or enteric infections. Intestinal epithelial luminal membrane NHE3 (Na+/H+ exchanger 3) and DRA (Down-Regulated in Adenoma; Cl-/HCO3- exchanger) play key roles in mediating electroneutral NaCl absorption. We have previously shown decreased NHE3 and DRA function in response to short-term infection with enteropathogenic E coli (EPEC), a diarrheal pathogen. Recent studies have also shown substantial downregulation of DRA expression in a diarrheal model of infection with Citrobacter rodentium, the mouse counterpart of EPEC. Since our previous studies showed that the probiotic Lactobacillus acidophilus (LA) increased DRA and NHE3 function and expression and conferred protective effects in experimental colitis, we sought to evaluate the efficacy of LA in counteracting NHE3 and DRA inhibition and ameliorating diarrhea in a model of C rodentium infection. FVB/N mice challenged with C rodentium [1 × 109 colony-forming units (CFU)] with or without administration of live LA (3 × 109 CFU) were assessed for NHE3 and DRA mRNA and protein expression, mRNA levels of carbonic anhydrase, diarrheal phenotype (assessed by colonic weight-to-length ratio), myeloperoxidase activity, and proinflammatory cytokines. LA counteracted C rodentium-induced inhibition of colonic DRA, NHE3, and carbonic anhydrase I and IV expression and attenuated diarrheal phenotype and MPO activity. Furthermore, LA completely blocked C rodentium induction of IL-1ß, IFN-γ, and CXCL1 mRNA and C rodentium-induced STAT3 phosphorylation. In conclusion, our data provide mechanistic insights into antidiarrheal effects of LA in a model of infectious diarrhea and colitis.


Subject(s)
Antiporters/metabolism , Citrobacter rodentium , Diarrhea/drug therapy , Enterobacteriaceae Infections/drug therapy , Lactobacillus acidophilus , Probiotics/therapeutic use , Sodium-Hydrogen Exchangers/metabolism , Animals , Colitis/metabolism , Colitis/microbiology , Colon/metabolism , Colon/microbiology , Cytokines/metabolism , Diarrhea/metabolism , Diarrhea/microbiology , Enterobacteriaceae Infections/metabolism , Enterobacteriaceae Infections/microbiology , Mice , Phosphorylation , Sodium-Hydrogen Exchanger 3 , Sulfate Transporters , Treatment Outcome
9.
Stem Cells ; 32(6): 1538-52, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24496925

ABSTRACT

Endothelial cell (EC) dedifferentiation in relation to neovascularization is a poorly understood process. In this report, we addressed the role of Wnt signaling in the mechanisms of neovascularization in adult tissues. Here, we show that a low-dose of 6-bromoindirubin-3'-oxime (BIO), a competitive inhibitor of glycogen synthase kinase-3ß, induced the stabilization of ß-catenin and its subsequent direct interaction with the transcription factor NANOG in the nucleus of ECs. This event induced loss of VE-cadherin from the adherens junctions, increased EC proliferation accompanied by asymmetric cell division (ACD), and formed cellular aggregates in hanging drop assays indicating the acquisition of a dedifferentiated state. In a chromatin immunoprecipitation assay, nuclear NANOG protein bound to the NANOG- and VEGFR2-promoters in ECs, and the addition of BIO activated the NANOG-promoter-luciferase reporter system in a cell-based assay. Consequently, NANOG-knockdown decreased BIO-induced NOTCH-1 expression, thereby decreasing cell proliferation, ACD, and neovascularization. In a Matrigel plug assay, BIO induced increased neovascularization, secondary to the presence of vascular endothelial growth factor (VEGF). Moreover, in a mouse model of hind limb ischemia, BIO augmented neovascularization that was coupled with increased expression of NOTCH-1 in ECs and increased smooth muscle α-actin(+) cell recruitment around the neovessels. Thus, these results demonstrate the ability of a low-dose of BIO to augment neovascularization secondary to VEGF, a process that was accompanied by a partial dedifferentiation of ECs via ß-catenin and the NANOG signaling pathway.


Subject(s)
Cell Dedifferentiation/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Indoles/pharmacology , Neovascularization, Physiologic/drug effects , Oximes/pharmacology , Angiogenesis Inducing Agents/metabolism , Animals , Cell Aggregation/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Fetal Proteins/genetics , Hindlimb/blood supply , Hindlimb/pathology , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Indoles/administration & dosage , Ischemia/pathology , Mice , Nanog Homeobox Protein , Oximes/administration & dosage , Phenotype , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Stability/drug effects , T-Box Domain Proteins/genetics , Vascular Endothelial Growth Factor A/pharmacology , beta Catenin/metabolism
10.
Blood ; 117(5): 1761-9, 2011 Feb 03.
Article in English | MEDLINE | ID: mdl-21119109

ABSTRACT

NANOG is a master transcription factor associated with the maintenance of stem cell pluripotency. Here, we demonstrate that transcription factor NANOG is expressed in cultured endothelial cells (ECs) and in a subset of tumor cell lines. Importantly, we provide evidence that WNT3A stimulation of ECs induces the transcription of NANOG which mediates the expression of vascular endothelial growth factor receptor-2, also known as fetal liver kinase-1 (FLK1). We defined ATTA as a minimal binding site for NANOG. Accordingly, a luciferase reporter assay showed that NANOG binds to and activates 4 ATTA binding sites identified in the FLK1 promoter after WNT3A stimulation. Consistent with this data, we found that, under basal conditions and in response to WNT3A stimulation, NANOG binding to these ATTA sequences markedly induced the expression of FLK1. Thus, our data indicate an essential role in angiogenesis for NANOG binding to these 4 ATTA sites. Surprisingly, NANOG depletion not only decreased FLK1 expression but also reduced cell proliferation and angiogenesis. These findings show the necessary and sufficient role of NANOG in inducing the transcription of FLK1 to regulate the angiogenic phenotypes of ECs.


Subject(s)
Cell Proliferation , Endothelium, Vascular/cytology , Homeodomain Proteins/metabolism , Neovascularization, Physiologic , Transcription, Genetic , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Dermis/cytology , Dermis/metabolism , Electrophoretic Mobility Shift Assay , Embryo, Mammalian , Endothelium, Vascular/metabolism , Fibroblasts , Flow Cytometry , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Immunoenzyme Techniques , Luciferases/metabolism , Mice , Nanog Homeobox Protein , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wnt Proteins , Wnt3 Protein , Wnt3A Protein
11.
Sci Transl Med ; 15(678): eabo0205, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36630481

ABSTRACT

The common γ chain (γc; IL-2RG) is a subunit of the interleukin (IL) receptors for the γc cytokines IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21. The lack of appropriate neutralizing antibodies recognizing IL-2RG has made it difficult to thoroughly interrogate the role of γc cytokines in inflammatory and autoimmune disease settings. Here, we generated a γc cytokine receptor antibody, REGN7257, to determine whether γc cytokines might be targeted for T cell-mediated disease prevention and treatment. Biochemical, structural, and in vitro analysis showed that REGN7257 binds with high affinity to IL-2RG and potently blocks signaling of all γc cytokines. In nonhuman primates, REGN7257 efficiently suppressed T cells without affecting granulocytes, platelets, or red blood cells. Using REGN7257, we showed that γc cytokines drive T cell-mediated disease in mouse models of graft-versus-host disease (GVHD) and multiple sclerosis by affecting multiple aspects of the pathogenic response. We found that our xenogeneic GVHD mouse model recapitulates hallmarks of acute and chronic GVHD, with T cell expansion/infiltration into tissues and liver fibrosis, as well as hallmarks of immune aplastic anemia, with bone marrow aplasia and peripheral cytopenia. Our findings indicate that γc cytokines contribute to GVHD and aplastic anemia pathology by promoting these characteristic features. By demonstrating that broad inhibition of γc cytokine signaling with REGN7257 protects from immune-mediated disorders, our data provide evidence of γc cytokines as key drivers of pathogenic T cell responses, offering a potential strategy for the management of T cell-mediated diseases.


Subject(s)
Anemia, Aplastic , Graft vs Host Disease , Interleukin Receptor Common gamma Subunit , T-Lymphocytes , Animals , Mice , Anemia, Aplastic/metabolism , Antibodies, Monoclonal/metabolism , Cytokines/metabolism , Graft vs Host Disease/metabolism , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Interleukin Receptor Common gamma Subunit/antagonists & inhibitors , Interleukin Receptor Common gamma Subunit/metabolism , Primates
12.
Microvasc Res ; 83(1): 64-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21616084

ABSTRACT

In this review, we discuss the role of focal adhesion kinase (FAK), an intracellular tyrosine kinase, in endothelial cells in relation to neovascularization. Genetic and in vitro studies have identified critical factors, receptor systems, and their intracellular signaling components that regulate the neovasculogenic phenotypes of endothelial cells. Among these factors, FAK appears to regulate several aspects of endothelial cellular behavior, including migration, survival, cytoskeletal organization, as well as cell proliferation. Upon adhesion of endothelial cells to extracellular matrix (ECM) ligands, integrins cluster on the plane of plasma-membrane, while cytoplasmic domains of integrins interact with cytoskeletal proteins and signaling molecules including FAK. However, FAK not only serves as a critical component of integrin signaling, but is also a downstream element of the VEGF/VEGF-receptor and other ligand-receptor systems that regulate neovascularization. A complete understanding of FAK-mediated neovascularization, therefore, should address the molecular and cellular mechanisms that regulate the biology of FAK. Continued research on FAK may, therefore, yield novel therapies to improve treatment modalities for the pathological neovascularization associated with diseases.


Subject(s)
Endothelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/enzymology , Neoplasms/blood supply , Neovascularization, Pathologic/enzymology , Neovascularization, Physiologic , Animals , Cell Differentiation , Cell Movement , Cell Proliferation , Endothelial Cells/pathology , Focal Adhesions/pathology , Humans , Neovascularization, Pathologic/pathology , Signal Transduction
13.
Mol Cancer ; 10: 51, 2011 May 11.
Article in English | MEDLINE | ID: mdl-21569306

ABSTRACT

BACKGROUND: The acquisition of proliferative and invasive phenotypes is considered a hallmark of neoplastic transformation; however, the underlying mechanisms are less well known. Lipid phosphate phosphatase-3 (LPP3) not only catalyzes the dephosphorylation of the bioactive lipid sphingosine-1-phosphate (S1P) to generate sphingosine but also may regulate embryonic development and angiogenesis via the Wnt pathway. The goal of this study was to determine the role of LPP3 in tumor cells. RESULTS: We observed increased expression of LPP3 in glioblastoma primary tumors and in U87 and U118 glioblastoma cell lines. We demonstrate that LPP3-knockdown inhibited both U87 and U118 glioblastoma cell proliferation in culture and tumor growth in xenograft assays. Biochemical experiments provided evidence that LPP3-knockdown reduced ß-catenin, CYCLIN-D1, and CD133 expression, with a concomitant increase in phosphorylated ß-catenin. In a converse experiment, the forced expression of LPP3 in human colon tumor (SW480) cells potentiated tumor growth via increased ß-catenin stability and CYCLIN-D1 synthesis. In contrast, elevated expression of LPP3 had no tumorigenic effects on primary cells. CONCLUSIONS: These results demonstrate for the first time an unexpected role of LPP3 in regulating glioblastoma progression by amplifying ß-catenin and CYCLIN-D1 activities.


Subject(s)
Cyclin D1/metabolism , Neoplasms/enzymology , Phosphatidate Phosphatase/metabolism , Signal Transduction , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Mice , Mice, Nude , Neoplasms/physiopathology , Phosphatidate Phosphatase/genetics , Tumor Burden , Xenograft Model Antitumor Assays
14.
J Crohns Colitis ; 15(10): 1720-1736, 2021 10 07.
Article in English | MEDLINE | ID: mdl-33690841

ABSTRACT

BACKGROUND AND AIMS: Dysfunction of the vitamin D receptor [VDR] contributes to the aetiology of IBD by regulating autophagy, immune response, and mucosal permeability. VDR directly controls the paracellular tight junction protein Claudin-2. Claudin-2 and Claudin-15 are unique in maintaining paracellular permeability. Interestingly, claudin-15 mRNA was downregulated in patients with ulcerative colitis. However, the exact mechanism of Claudin-15 regulation in colitis is still unknown. Here, we investigated the protective role of VDR against intestinal inflammation via upregulating Claudin-15. METHODS: We analysed the correlation of Claudin-15 with the reduction of VDR in human colitis. We generated intestinal epithelial overexpression of VDR [O-VDR] mice to study the gain of function of VDR in colitis. Intestinal epithelial VDR knockout [VDR∆IEC] mice were used for the loss of function study. Colonoids and SKCO15 cells were used as in vitro models. RESULTS: Reduced Claudin-15 was significantly correlated with decreased VDR along the colonic epithelium of human IBD. O-VDR mice showed decreased susceptibility to chemically and bacterially induced colitis and marked increased Claudin-15 expression [both mRNA and protein] in the colon. Correspondingly, colonic Claudin-15 was reduced in VDR∆IEC mice, which were susceptible to colitis. Overexpression of intestinal epithelial VDR and vitamin D treatment resulted in a significantly increased Claudin-15. ChIP assays identified the direct binding of VDR to the claudin-15 promoter, suggesting that claudin-15 is a target gene of VDR. CONCLUSION: We demonstrated the mechanism of VDR upregulation of Claudin-15 to protect against colitis. This might enlighten the mechanism of barrier dysfunction in IBD and potential therapeutic strategies to inhibit inflammation.


Subject(s)
Claudins/metabolism , Colitis/prevention & control , Intestinal Mucosa/metabolism , Receptors, Calcitriol/metabolism , Aged , Animals , Claudins/genetics , Colon/metabolism , Female , Humans , Male , Mice, Knockout , Middle Aged , RNA, Messenger/metabolism , Up-Regulation
15.
J Immunother Cancer ; 9(1)2021 01.
Article in English | MEDLINE | ID: mdl-33483343

ABSTRACT

BACKGROUND: Programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) blocking antibodies including cemiplimab have generated profound clinical activity across diverse cancer types. Tumorous PD-L1 expression, as assessed by immunohistochemistry (IHC), is an accepted predictive marker of response to therapy in some cancers. However, expression is often dynamic and heterogeneous, and therefore not reliably captured by IHC from tumor biopsies or archival samples. Thus, there is significant need for accurate whole-body quantification of PD-L1 levels. METHODS: We radiolabeled the novel human anti-PD-L1 antibody REGN3504 with zirconium-89 (89Zr) using the chelator p-SCN-Bn-Deferoxamine to enable non-invasive immuno-positron emission tomography (immuno-PET) of PD-L1 expression. PET imaging assessed the localization of 89Zr-REGN3504 to multiple human tumor xenografts. Mice genetically humanized for PD-1 and PD-L1 were used to assess the biodistribution of 89Zr-REGN3504 to normal tissues and the estimated human radiation dosimetry of 89Zr-REGN3504 was also determined. Pharmacokinetics of REGN3504 was assessed in monkeys. RESULTS: Clear localization of 89Zr-REGN3504 to human tumor xenografts was observed via PET imaging and ex vivo biodistribution studies demonstrated high (fourfold to sixfold) tumor:blood ratios. 89Zr-REGN3504 specifically localized to spleen and lymph nodes in the PD-1/PD-L1 humanized mice. 89Zr-REGN3504 immuno-PET accurately detected a significant reduction in splenic PD-L1 positive cells following systemic treatment with clodronate liposomes. Radiation dosimetry suggested absorbed doses would be within guidelines for other 89Zr radiolabeled, clinically used antibodies. Pharmacokinetics of REGN3504 was linear. CONCLUSION: This work supports the clinical translation of 89Zr-REGN3504 immuno-PET for the assessment of PD-L1 expression. Future clinical studies will aim to investigate the utility of 89Zr-REGN3504 immuno-PET for predicting and monitoring response to anti-PD-1 therapy.


Subject(s)
Antibodies, Monoclonal/administration & dosage , B7-H1 Antigen/metabolism , Neoplasms/diagnostic imaging , Radioisotopes/chemistry , Zirconium/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacokinetics , Case-Control Studies , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Haplorhini , Humans , Male , Mice , Neoplasm Transplantation , Neoplasms/immunology , Positron-Emission Tomography , Tissue Distribution
16.
Sci Rep ; 10(1): 7340, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32355205

ABSTRACT

Microbiota derived metabolites act as chemical messengers that elicit a profound impact on host physiology. Vitamin D receptor (VDR) is a key genetic factor for shaping the host microbiome. However, it remains unclear how microbial metabolites are altered in the absence of VDR. We investigated metabolites from mice with tissue-specific deletion of VDR in intestinal epithelial cells or myeloid cells. Conditional VDR deletion severely changed metabolites specifically produced from carbohydrate, protein, lipid, and bile acid metabolism. Eighty-four out of 765 biochemicals were significantly altered due to the Vdr status, and 530 significant changes were due to the high-fat diet intervention. The impact of diet was more prominent due to loss of VDR as indicated by the differences in metabolites generated from energy expenditure, tri-carboxylic acid cycle, tocopherol, polyamine metabolism, and bile acids. The effect of HFD was more pronounced in female mice after VDR deletion. Interestingly, the expression levels of farnesoid X receptor in liver and intestine were significantly increased after intestinal epithelial VDR deletion and were further increased by the high-fat diet. Our study highlights the gender differences, tissue specificity, and potential gut-liver-microbiome axis mediated by VDR that might trigger downstream metabolic disorders.


Subject(s)
Gastrointestinal Microbiome , Myeloid Cells/microbiology , Receptors, Calcitriol/genetics , Animals , Bile Acids and Salts/metabolism , Carbohydrate Metabolism , Diet, High-Fat , Energy Metabolism , Female , Gene Deletion , Intestines , Lipid Metabolism/genetics , Liver/metabolism , Male , Metabolic Syndrome/genetics , Mice , Mice, Knockout , Polyamines/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Sex Factors , Signal Transduction , Tocopherols/metabolism
17.
Cell Mol Gastroenterol Hepatol ; 10(4): 729-746, 2020.
Article in English | MEDLINE | ID: mdl-32497792

ABSTRACT

BACKGROUND & AIMS: Vitamin D exerts regulatory roles via vitamin D receptor (VDR) in mucosal immunity, host defense, and inflammation involving host factors and microbiome. Human Vdr gene variation shapes the microbiome and VDR deletion leads to dysbiosis. Low VDR expression and diminished vitamin D/VDR signaling are observed in colon cancer. Nevertheless, how intestinal epithelial VDR is involved in tumorigenesis through gut microbiota remains unknown. We hypothesized that intestinal VDR protects mice against dysbiosis via modulating the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway in tumorigenesis. METHODS: To test our hypothesis, we used an azoxymethane/dextran sulfate sodium-induced cancer model in intestinal VDR conditional knockout (VDRΔIEC) mice, cell cultures, stem cell-derived colonoids, and human colon cancer samples. RESULTS: VDRΔIEC mice have higher numbers of tumors, with the location shifted from the distal to proximal colon. Fecal microbiota analysis showed that VDR deletion leads to a bacterial profile shift from normal to susceptible carcinogenesis. We found enhanced bacterial staining in mouse and human tumors. Microbial metabolites from VDRΔIEC mice showed increased secondary bile acids, consistent with observations in human CRC. We further identified that VDR protein bound to the Jak2 promoter, suggesting that VDR transcriptionally regulated Jak2. The JAK/STAT pathway is critical in intestinal and microbial homeostasis. Fecal samples from VDRΔIEC mice activate the STAT3 signaling in human and mouse organoids. Lack of VDR led to hyperfunction of Jak2 in response to intestinal dysbiosis. A JAK/STAT inhibitor abolished the microbiome-induced activation of STAT3. CONCLUSIONS: We provide insights into the mechanism of VDR dysfunction leading to dysbiosis and tumorigenesis. It indicates a new target: microbiome and VDR for the prevention of cancer.


Subject(s)
Carcinogenesis/metabolism , Colonic Neoplasms/metabolism , Dysbiosis/metabolism , Receptors, Calcitriol/metabolism , Aged , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Dysbiosis/genetics , Dysbiosis/pathology , Female , HCT116 Cells , Humans , Intestines/pathology , Janus Kinases/metabolism , Male , Mice, Inbred C57BL , Middle Aged , Protective Factors , Receptors, Calcitriol/genetics , STAT Transcription Factors/metabolism , Signal Transduction
18.
Inflamm Bowel Dis ; 26(4): 534-545, 2020 03 04.
Article in English | MEDLINE | ID: mdl-31634391

ABSTRACT

BACKGROUND: Intestinal epithelial apical membrane Cl-/HCO3- exchanger DRA (downregulated in adenoma, SLC26A3) has emerged as an important therapeutic target for diarrhea, emphasizing the potential therapeutic role of agents that upregulate DRA. All-trans retinoic acid (ATRA), a key vitamin A metabolite, was earlier shown by us to stimulate DRA expression in intestinal epithelial cells. However, its role in modulating DRA in gut inflammation has not been investigated. AIMS: Our aim was to analyze the efficacy of ATRA in counteracting inflammation-induced decrease in DRA in vitro and in vivo. METHODS: Interferon-γ (IFN-γ)-treated Caco-2 cells and dextran sulfate sodium (DSS)-treated C57BL/6J mice served as in vitro and in vivo models of gut inflammation, respectively. The effect of ATRA on IFN-γ-mediated inhibition of DRA function, expression, and promoter activity were elucidated. In the DSS colitis model, diarrheal phenotype, cytokine response, in vivo imaging, myeloperoxidase activity, and DRA expression were measured in the distal colon. RESULTS: All-trans retinoic acid (10 µM, 24 h) abrogated IFN-γ (30 ng/mL, 24 h)-induced decrease in DRA function, expression, and promoter activity in Caco-2 cells. All-trans retinoic acid altered IFN-γ signaling via blocking IFN-γ-induced tyrosine phosphorylation of STAT-1. All-trans retinoic acid cotreatment (1 mg/kg BW, i.p. daily) of DSS-treated mice (3% in drinking water for 7 days) alleviated colitis-associated weight loss, diarrheal phenotype, and induction of IL-1ß and CXCL1 and a decrease in DRA mRNA and protein levels in the colon. CONCLUSION: Our data showing upregulation of DRA under normal and inflammatory conditions by ATRA demonstrate a novel role of this micronutrient in alleviating IBD-associated diarrhea.


Subject(s)
Antiporters/metabolism , Chloride-Bicarbonate Antiporters/metabolism , Colitis/drug therapy , Intestinal Mucosa/drug effects , Sulfate Transporters/metabolism , Tretinoin/pharmacology , Animals , Antiporters/genetics , Caco-2 Cells , Chloride-Bicarbonate Antiporters/genetics , Colon/metabolism , Dextran Sulfate/toxicity , Diarrhea/drug therapy , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Inflammation/metabolism , Interferon-gamma/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , Sulfate Transporters/genetics , Up-Regulation , Weight Loss/drug effects
19.
Article in English | MEDLINE | ID: mdl-30828578

ABSTRACT

Metabolic syndrome is a multi-faceted disease. The microbiota, as a newly discovered organ, contributes to the pathogenesis and progression of metabolic syndrome. Recent studies have demonstrated that nuclear receptors play critical roles in metabolic diseases. In the current review, we discuss the general role of the microbiome in health and metabolic syndrome. We summarize the functions of the nuclear receptor vitamin D receptor (VDR) in metabolism. The focus of this review is the novel roles of vitamin D/VDR signaling in regulating inflammation and the microbiome, especially in obesity. Furthermore, we extend our discussion of potential gut-liver axis mediated by VDR signaling and microbiota in obesity. Finally, we discuss the potential clinical application of probiotics and fecal microbiota transplantation in prevention and treatment of metabolic syndrome. Insights into nuclear receptors in metabolism and metabolic diseases will allow us to develop new strategies for fighting metabolic diseases.

20.
J Vis Exp ; (121)2017 03 16.
Article in English | MEDLINE | ID: mdl-28362399

ABSTRACT

The intestinal epithelium has important transport and barrier functions that play key roles in normal physiological functions of the body while providing a barrier to foreign particles. Impaired epithelial transport (ion, nutrient, or drugs) has been associated with many diseases and can have consequences that extend beyond the normal physiological functions of the transporters, such as by influencing epithelial integrity and the gut microbiome. Understanding the function and regulation of transport proteins is critical for the development of improved therapeutic interventions. The biggest challenge in the study of epithelial transport is developing a suitable model system that recapitulates important features of the native intestinal epithelial cells. Several in vitro cell culture models, such as Caco-2, T-84, and HT-29-Cl.19A cells are typically used in epithelial transport research. These cell lines represent a reductionist approach to modeling the epithelium and have been used in many mechanistic studies, including their examination of epithelial-microbial interactions. However, cell monolayers do not accurately reflect cell-cell interactions and the in vivo microenvironment. Cells grown in 3D have shown to be promising models for drug permeability studies. We show that Caco-2 cells in 3D can be used to study epithelial transporters. It is also important that studies in Caco-2 cells are complemented with other models to rule out cell specific effects and to take into account the complexity of the native intestine. Several methods have been previously used to assess the functionality of transporters, such as everted sac and uptake in isolated epithelial cells or in isolated plasma membrane vesicles. Taking into consideration the challenges in the field with respect to models and the measurement of transport function, we demonstrate here a protocol to grow Caco-2 cells in 3D and describe the use of an Ussing chamber as an effective approach to measure serotonin transport, such as in intact polarized intestinal epithelia.


Subject(s)
Carrier Proteins/metabolism , Intestinal Mucosa/metabolism , Biological Transport , Caco-2 Cells/metabolism , Cell Culture Techniques , Cell Membrane/metabolism , Cell Membrane Permeability , Epithelial Cells/metabolism , Epithelium/metabolism , Humans , Intestinal Mucosa/cytology , Intestines/cytology
SELECTION OF CITATIONS
SEARCH DETAIL