Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 131(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34128475

ABSTRACT

FOXP3+ Tregs are expanded within the inflamed intestine of human Crohn's disease, yet FOXP3-mediated gene repression within these cells is lost. The polycomb repressive complexes play a role in FOXP3 target gene regulation, but deeper mechanistic insight is incomplete. We have now specifically identified the polycomb-repressive complex 1 (PRC1) family member, BMI1 in the regulation of a proinflammatory enhancer network in both human and murine Tregs. Using human Tregs and lamina propria T cells, we inferred PRC1 to regulate Crohn's associated gene networks through assays of chromatin accessibility. Conditional deletion of BMI1 in murine FOXP3+ cells led to systemic inflammation. BMI1-deficient Tregs beared a TH1/TH17-like phenotype as assessed by assays of genome wide transcription, chromatin accessibility and proteomic techniques. Finally, BMI1 mutant FOXP3+ cells did not suppress colitis in the adoptive transfer model of human inflammatory bowel disease. We propose that BMI1 plays an important role in enforcing Treg identity in vitro and in vivo. Loss of Treg identity via genetic or transient BMI1 depletion perturbs the epigenome and converts Tregs into Th1/Th17-like proinflammatory cells, a transition relevant to human Crohn's disease associated CD4+ T cells.


Subject(s)
Crohn Disease/immunology , Epigenesis, Genetic/immunology , Polycomb Repressive Complex 1/immunology , Proto-Oncogene Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Crohn Disease/genetics , Humans , Mice , Mice, Transgenic , Polycomb Repressive Complex 1/genetics , Proto-Oncogene Proteins/genetics , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th17 Cells/immunology
2.
J Crohns Colitis ; 14(1): 96-109, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31158273

ABSTRACT

BACKGROUND: The aetiology of Crohn's disease [CD] involves immune dysregulation in a genetically susceptible individual. Genome-wide association studies [GWAS] have identified 200 loci associated with CD, ulcerative colitis, or both, most of which fall within non-coding DNA regions. Long non-coding RNAs [lncRNAs] regulate gene expression by diverse mechanisms and have been associated with disease activity in inflammatory bowel disease. However, disease-associated lncRNAs have not been characterised in pathogenic immune cell populations. METHODS: Terminal ileal samples were obtained from 22 CD patients and 13 controls. RNA from lamina propria CD4+ T cells was sequenced and long intergenic non-coding RNAs [lincRNAs] were detected. Overall expression patterns, differential expression [DE], and pathway and gene enrichment analyses were performed. Knockdown of novel lincRNAs XLOC_000261 and XLOC_000014 was performed. Expression of Th1 or Th17-associated transcription factors, T-bet and RORγt, respectively, was assessed by flow cytometry. RESULTS: A total of 6402 lincRNAs were expressed, 960 of which were novel. Unsupervised clustering and principal component analysis showed that the lincRNA expression discriminated patients from controls. A total of 1792 lincRNAs were DE, and 295 [79 novel; 216 known] mapped to 267 of 5727 DE protein-coding genes. The novel lincRNAs were enriched in inflammatory and Notch signalling pathways [p <0.05]. Furthermore, DE lincRNAs in CD patients were more frequently found in DNA regions with known inflammatory bowel disease [IBD]-associated loci. The novel lincRNA XLOC_000261 negatively regulated RORγt expression in Th17 cells. CONCLUSIONS: We describe a novel set of DE lincRNAs in CD-associated CD4+ cells and demonstrate that novel lincRNA XLOC_000261 appears to negatively regulate RORγt protein expression in Th17 cells.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Crohn Disease/etiology , RNA, Long Noncoding/metabolism , Aged , Case-Control Studies , Crohn Disease/metabolism , Crohn Disease/pathology , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Mucous Membrane/metabolism
3.
Article in English | MEDLINE | ID: mdl-30510991

ABSTRACT

Background & Aims: Forkhead box protein 3 (FOXP3)+ regulatory T cell (Treg) dysfunction is associated with autoimmune diseases; however, the mechanisms responsible for inflammatory bowel disease pathophysiology are poorly understood. Here, we tested the hypothesis that a physical interaction between transcription factor FOXP3 and the epigenetic enzyme enhancer of zeste homolog 2 (EZH2) is essential for gene co-repressive function. Methods: Human FOXP3 mutations clinically relevant to intestinal inflammation were generated by site-directed mutagenesis. T lymphocytes were isolated from mice, human blood, and lamina propria of Crohn's disease (CD) patients and non-CD controls. We performed proximity ligation or a co-immunoprecipitation assay in FOXP3-mutant+, interleukin 6 (IL6)-treated or CD-CD4+ T cells to assess FOXP3-EZH2 protein interaction. We studied IL2 promoter activity and chromatin state of the interferon γ locus via luciferase reporter and chromatin-immunoprecipitation assays, respectively, in cells expressing FOXP3 mutants. Results: EZH2 binding was abrogated by inflammatory bowel disease-associated FOXP3 cysteine 232 (C232) mutation. The C232 mutant showed impaired repression of IL2 and diminished EZH2-mediated trimethylation of histone 3 at lysine 27 on interferon γ, indicative of compromised Treg physiologic function. Generalizing this mechanism, IL6 impaired FOXP3-EZH2 interaction. IL6-induced effects were reversed by Janus kinase 1/2 inhibition. In lamina propria-derived CD4+T cells from CD patients, we observed decreased FOXP3-EZH2 interaction. Conclusions: FOXP3-C232 mutation disrupts EZH2 recruitment and gene co-repressive function. The proinflammatory cytokine IL6 abrogates FOXP3-EZH2 interaction. Studies in lesion-derived CD4+ T cells have shown that reduced FOXP3-EZH2 interaction is a molecular feature of CD patients. Destabilized FOXP3-EZH2 protein interaction via diverse mechanisms and consequent Treg abnormality may drive gastrointestinal inflammation.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Forkhead Transcription Factors/metabolism , Inflammation/metabolism , Inflammation/pathology , Intestines/pathology , Adult , Animals , Cell Nucleus/metabolism , Cell Separation , Co-Repressor Proteins/metabolism , Female , Humans , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Interleukin-6/metabolism , Janus Kinases/metabolism , Jurkat Cells , Male , Mice, Inbred C57BL , Middle Aged , Mutation/genetics , Phosphorylation , Phosphotyrosine/metabolism , Polycomb Repressive Complex 2/metabolism , Protein Binding , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism
4.
J Biol Chem ; 292(2): 706-722, 2017 Jan 13.
Article in English | MEDLINE | ID: mdl-27909059

ABSTRACT

Regulatory T (Treg) cells expressing the transcription factor FOXP3 play a pivotal role in maintaining immunologic self-tolerance. We and others have shown previously that EZH2 is recruited to the FOXP3 promoter and its targets in Treg cells. To further address the role for EZH2 in Treg cellular function, we have now generated mice that lack EZH2 specifically in Treg cells (EZH2Δ/ΔFOXP3+). We find that EZH2 deficiency in FOXP3+ T cells results in lethal multiorgan autoimmunity. We further demonstrate that EZH2Δ/ΔFOXP3+ T cells lack a regulatory phenotype in vitro and secrete proinflammatory cytokines. Of special interest, EZH2Δ/ΔFOXP3+ mice develop spontaneous inflammatory bowel disease. Guided by these results, we assessed the FOXP3 and EZH2 gene networks by RNA sequencing in isolated intestinal CD4+ T cells from patients with Crohn's disease. Gene network analysis demonstrates that these CD4+ T cells display a Th1/Th17-like phenotype with an enrichment of gene targets shared by FOXP3 and EZH2. Combined, these results suggest that the inflammatory milieu found in Crohn's disease could lead to or result from deregulation of FOXP3/EZH2-enforced T cell gene networks contributing to the underlying intestinal inflammation.


Subject(s)
Crohn Disease/immunology , Enhancer of Zeste Homolog 2 Protein/immunology , Gene Regulatory Networks/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Crohn Disease/pathology , Cytokines/genetics , Cytokines/immunology , Enhancer of Zeste Homolog 2 Protein/genetics , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Mice , Mice, Transgenic , T-Lymphocytes, Regulatory/pathology , Th17 Cells/pathology
5.
Am J Physiol Gastrointest Liver Physiol ; 309(11): G900-9, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26472224

ABSTRACT

Krüppel-like factor (KLF)-10 is an important transcriptional regulator of TGF-ß1 signaling in both CD8(+) and CD4(+) T lymphocytes. In the present study, we demonstrate a novel role for KLF10 in the regulation of TGFßRII expression with functional relevance in macrophage differentiation and activation. We first show that transfer of KLF10(-/-) bone marrow-derived macrophages into wild-type (WT) mice leads to exacerbation of experimental colitis. At the cell biological level, using two phenotypic strategies, we show that KLF10-deficient mice have an altered colonic macrophage phenotype with higher frequency of proinflammatory LyC6(+)MHCII(+) cells and a reciprocal decrease of the anti-inflammatory LyC6(-)MHCII(+) subset. Additionally, the anti-inflammatory CD11b(+)CX3CR1(hi) subset of colonic macrophages is significantly decreased in KLF10(-/-) compared with WT mice under inflammatory conditions. Molecularly, CD11b(+) colonic macrophages from KLF10(-/-) mice exhibit a proinflammatory cytokine profile with increased production of TNF-α and lower production of IL-10 in response to LPS stimulation. Because KLF10 is a transcription factor, we explored how this protein may regulate macrophage function. Consequently, we analyzed the expression of TGFßRII expression in colonic macrophages and found that, in the absence of KLF10, macrophages express lower levels of TGFßRII and display an attenuated Smad-2 phosphorylation following TGF-ß1 stimulation. We further show that KLF10 directly binds to the TGFßRII promoter in macrophages, leading to enhanced gene expression through histone H3 acetylation. Collectively, our data reveal a critical role for KLF10 in the epigenetic regulation of TGFßRII expression in macrophages and the acquisition of a "regulatory" phenotype that contributes to intestinal mucosal homeostasis.


Subject(s)
Colitis/metabolism , Colon/metabolism , Early Growth Response Transcription Factors/deficiency , Intestinal Mucosa/metabolism , Kruppel-Like Transcription Factors/deficiency , Macrophages/metabolism , Acetylation , Animals , Base Sequence , Binding Sites , CD11b Antigen/metabolism , CX3C Chemokine Receptor 1 , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Early Growth Response Transcription Factors/genetics , Genetic Predisposition to Disease , Histocompatibility Antigens Class II/metabolism , Histones/metabolism , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Intestinal Mucosa/pathology , Kruppel-Like Transcription Factors/genetics , Macrophages/transplantation , Mice, Knockout , Molecular Sequence Data , Phenotype , Phosphorylation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Chemokine/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism
6.
Cell Mol Gastroenterol Hepatol ; 1(2): 188-202.e4, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25750932

ABSTRACT

BACKGROUND AND AIMS: KLF proteins function as epigenetic reprogramming factors during cell differentiation in many cell populations and in engineered iPS cells. In this study, we determined KLF14 function in the regulation of FOXP3, a transcription factor critical for Treg cell differentiation. METHODS: We studied the effects of KLF14 on FOXP3 expression at the level of the protein and mRNA. We evaluated the functional relevance of KLF14 to FOXP3+ Treg cells in vitro and in vivo through suppression assays and two colitis models. Finally, we analyzed the effect of KLF14 on the epigenetic landscape of the FOXP3 promoter locus through chromatin immuno-precipitation. RESULTS: KLF14, induced upon activation of naïve CD4+ T cells, segregates to the FOXP3- population and is inversely associated with FOXP3 expression and Treg function. KLF14 KO CD4+ cells differentiated into adaptive Tregs more readily in vitro and in vivo. KLF14 KO cells demonstrated enhanced Treg suppressor function in vitro and in vivo. KLF14 repressed FOXP3 at the level of the mRNA and protein, and by ChIP assay KLF14 was found to bind to the TSDR enhancer region of FOXP3. Furthermore, loss of KLF14 reduced the levels of H3K9me3, HP1 and Suv39H1at the TSDR. CONCLUSIONS: These results outline a novel mechanism by which KLF14 regulates Treg cell differentiation via chromatin remodeling at the FOXP3 TSDR. To our knowledge, this is the first evidence supporting a role for KLF14 in maintaining the differentiated state of Treg cells and outlines a potential mechanism to modify the expression of immune genes, such as FOXP3, which are critical to T cell fate.

7.
Gastroenterology ; 148(5): 978-90, 2015 May.
Article in English | MEDLINE | ID: mdl-25637652

ABSTRACT

BACKGROUND & AIMS: After allogeneic transplantation, murine stem cells (SCs) for interstitial cells of Cajal (ICCs), electrical pacemaker, and neuromodulator cells of the gut, were incorporated into gastric ICC networks, indicating in vivo immunosuppression. Immunosuppression is characteristic of bone marrow- and other non-gut-derived mesenchymal stem cells (MSCs), which are emerging as potential therapeutic agents against autoimmune diseases, including inflammatory bowel disease. Therefore, we investigated whether gut-derived ICC-SCs could also mitigate experimental colitis and studied the mechanisms of ICC-SC-mediated immunosuppression in relation to MSC-induced pathways. METHODS: Isolated ICC-SCs were studied by transcriptome profiling, cytokine assays, flow cytometry, mixed lymphocyte reaction, and T-cell proliferation assay. Mice with acute and chronic colitis induced by dextran sulfate sodium and T-cell transfer, respectively, were administered ICC-SCs intraperitoneally and evaluated for disease activity by clinical and pathological assessment and for ICC-SC homing by live imaging. RESULTS: Unlike strain-matched dermal fibroblasts, intraperitoneally administered ICC-SCs preferentially homed to the colon and reduced the severity of both acute and chronic colitis assessed by clinical and blind pathological scoring. ICC-SCs profoundly suppressed T-cell proliferation in vitro. Similar to MSCs, ICC-SCs strongly expressed cyclooxygenase 1/2 and basally secreted prostaglandin E2. Indomethacin, a cyclooxygenase inhibitor, countered the ICC-SC-mediated suppression of T-cell proliferation. In contrast, we found no role for regulatory T-cell-, programmed death receptor-, and transforming growth factor-ß-mediated mechanisms reported in MSCs; and transcriptome profiling did not support a relationship between ICC-SCs and MSCs. CONCLUSIONS: Murine ICC-SCs belong to a class different from MSCs and potently mitigate experimental colitis via prostaglandin E2-mediated immunosuppression.


Subject(s)
Colitis/prevention & control , Colon , Dinoprostone/metabolism , Immunity, Cellular , Interstitial Cells of Cajal/transplantation , Stem Cell Transplantation , Adoptive Transfer , Animals , Cell Proliferation , Cells, Cultured , Colitis/chemically induced , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Colon/immunology , Colon/metabolism , Colon/pathology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dextran Sulfate , Gene Expression Profiling , Genetic Markers , Homeodomain Proteins/genetics , Immunocompromised Host , Interstitial Cells of Cajal/immunology , Interstitial Cells of Cajal/metabolism , Lymphocyte Activation , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Time Factors
8.
Am J Physiol Cell Physiol ; 308(5): C362-71, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25472963

ABSTRACT

KLF10 has recently elicited significant attention as a transcriptional regulator of transforming growth factor-ß1 (TGF-ß1) signaling in CD4(+) T cells. In the current study, we demonstrate a novel role for KLF10 in the regulation of TGF-ß receptor II (TGF-ßRII) expression with functional relevance in antiviral immune response. Specifically, we show that KLF10-deficient mice have an increased number of effector/memory CD8(+) T cells, display higher levels of the T helper type 1 cell-associated transcription factor T-bet, and produce more IFN-γ following in vitro stimulation. In addition, KLF10(-/-) CD8(+) T cells show enhanced proliferation in vitro and homeostatic proliferation in vivo. Freshly isolated CD8(+) T cells from the spleen of adult mice express lower levels of surface TGF-ßRII (TßRII). Congruently, in vitro activation of KLF10-deficient CD8(+) T cells upregulate TGF-ßRII to a lesser extent compared with wild-type (WT) CD8(+) T cells, which results in attenuated Smad2 phosphorylation following TGF-ß1 stimulation compared with WT CD8(+) T cells. Moreover, we demonstrate that KLF10 directly binds to the TGF-ßRII promoter in T cells, leading to enhanced gene expression. In vivo viral infection with Daniel's strain Theiler's murine encephalomyelitis virus (TMEV) also led to lower expression of TGF-ßRII among viral-specific KLF10(-/-) CD8(+) T cells and a higher percentage of IFN-γ-producing CD8(+) T cells in the spleen. Collectively, our data reveal a critical role for KLF10 in the transcriptional activation of TGF-ßRII in CD8(+) T cells. Thus, KLF10 regulation of TGF-ßRII in this cell subset may likely play a critical role in viral and tumor immune responses for which the integrity of the TGF-ß1/TGF-ßRII signaling pathway is crucial.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Early Growth Response Transcription Factors/physiology , Kruppel-Like Transcription Factors/physiology , Protein Serine-Threonine Kinases/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/biosynthesis , Animals , Cells, Cultured , Early Growth Response Transcription Factors/deficiency , Gene Expression Regulation , Humans , Jurkat Cells , Kruppel-Like Transcription Factors/deficiency , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptor, Transforming Growth Factor-beta Type II
9.
Am J Physiol Regul Integr Comp Physiol ; 307(6): R608-20, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24944246

ABSTRACT

Inducible gene expression, which requires chromatin remodeling on gene promoters, underlies the epigenetically inherited differentiation program of most immune cells. However, chromatin-mediated mechanisms that underlie these events in T regulatory cells remain to be fully characterized. Here, we report that inducibility of FOXP3, a key transcription factor for the development of T regulatory cells, depends upon Kruppel-like factor 10 (KLF10) interacting with two antagonistic histone-modifying systems. We utilized chromatin immunoprecipitation, genome-integrated reporter assays, and functional domain KLF10 mutant proteins, to characterize reciprocal interactions between this transcription factor and either the Sin3-histone deacetylase complex or the histone acetyltransferase, p300/CBP-associated factor (PCAF). We characterize a Sin3-interacting repressor domain on the NH2 terminus of KLF10, which works to limit the activating function of this transcription factor. Indeed, inactivation of this Sin3-interacting domain renders KLF10 able to physically associate with PCAF as to induce FOXP3 gene transcription. We show that this biochemical data derived from studying our genome-integrated reporter cell system are recapitulated in primary murine lymphocytes. Collectively, these results advance our understanding of how a single transcription factor, namely KLF10, functions as a toggle to integrate antagonistic signals regulating FOXP3 and, thus, immune activation.


Subject(s)
Colitis/enzymology , Colon/enzymology , Early Growth Response Transcription Factors/metabolism , Forkhead Transcription Factors/metabolism , Kruppel-Like Transcription Factors/metabolism , Sin3 Histone Deacetylase and Corepressor Complex/metabolism , T-Lymphocytes, Regulatory/enzymology , p300-CBP Transcription Factors/metabolism , Animals , Binding Sites , Chromatin Assembly and Disassembly , Colitis/chemically induced , Colitis/genetics , Colitis/immunology , Colon/immunology , Dextran Sulfate , Disease Models, Animal , Early Growth Response Transcription Factors/chemistry , Early Growth Response Transcription Factors/deficiency , Early Growth Response Transcription Factors/genetics , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Humans , Jurkat Cells , Kruppel-Like Transcription Factors/chemistry , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Models, Molecular , Mutation , Promoter Regions, Genetic , Protein Conformation , Protein Interaction Domains and Motifs , Signal Transduction , Sin3 Histone Deacetylase and Corepressor Complex/chemistry , T-Lymphocytes, Regulatory/immunology , Transfection , Up-Regulation
10.
J Pediatr Gastroenterol Nutr ; 58(5): 561-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24792626

ABSTRACT

OBJECTIVES: Inflammatory bowel disease (IBD) is heritable, but a total of 163 variants commonly implicated in IBD pathogenesis account for only 25% of the heritability. Rare, highly penetrant genetic variants may also explain mendelian forms of IBD and some of the missing heritability. To test the hypothesis that rare loss-of-function mutations can be causative, we performed whole exome sequencing (WES) on 5 members of a 2-generation family of European ancestry presenting with an early-onset and atypical form of IBD. METHODS: WES was performed for all of the 5 family members; the mother and 3 male offspring were affected, whereas the father was unaffected. Mapping, annotation, and filtering criteria were used to reduce candidate variants. For functional testing we performed forkhead box P3 (FOXP3) staining and a T-cell suppression assay. RESULTS: We identified a novel missense variant in exon 6 of the X-linked FOXP3 gene. The c.694A>C substitution in FOXP3 results in a cysteine-to-glycine change at the protein position 232 that is completely conserved among all vertebrates. This variant (heterozygous in the mother and hemizygous in all 3 affected sons) did not impair FOXP3 protein expression, but significantly reduced the ability of the host's T regulatory cells to suppress an inappropriate autoimmune response. The variant results in a milder immune dysregulation, polyendocrinopathy, enteropathy, and X-linked phenotype with early-onset IBD. CONCLUSIONS: Our study illustrates the successful application of WES for making a definitive molecular diagnosis in a case of multiply affected families, with atypical IBD-like phenotype. Our results also have important implications for disease biology and disease-directed therapeutic development.


Subject(s)
Exome/genetics , Forkhead Transcription Factors/genetics , Inflammatory Bowel Diseases/genetics , Mutation , Eczema/genetics , Female , Forkhead Transcription Factors/analysis , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/immunology , Genotype , Humans , Infant , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/pathology , Male , Mutation, Missense/genetics , Pedigree , Phenotype , Polyendocrinopathies, Autoimmune/genetics , Sequence Analysis, DNA , T-Lymphocytes, Regulatory/immunology
11.
J Biol Chem ; 289(23): 16516-25, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24737325

ABSTRACT

The role of GLI1 in pancreatic tumor initiation promoting the progression of preneoplastic lesions into tumors is well established. However, its function at later stages of pancreatic carcinogenesis remains poorly understood. To address this issue, we crossed the gli1 knock-out (GKO) animal with cre-dependent pancreatic activation of oncogenic kras concomitant with loss of the tumor suppressor tp53 (KPC). Interestingly, in this model, GLI1 played a tumor-protective function, where survival of GKO/KPC mice was reduced compared with KPC littermates. Both cohorts developed pancreatic cancer without significant histopathological differences in survival studies. However, analysis of mice using ultrasound-based imaging at earlier time points showed increased tumor burden in GKO/KPC mice. These animals have larger tumors, decreased body weight, increased lactate dehydrogenase production, and severe leukopenia. In vivo and in vitro expression studies identified FAS and FAS ligand (FASL) as potential mediators of this phenomenon. The FAS/FASL axis, an apoptotic inducer, plays a role in the progression of pancreatic cancer, where its expression is usually lost or significantly reduced in advanced stages of the disease. Chromatin immunoprecipitation and reporter assays identified FAS and FASL as direct targets of GLI1, whereas GKO/KPC mice showed lower levels of this ligand compared with KPC animals. Finally, decreased levels of apoptosis were detected in tumor tissue in the absence of GLI1 by TUNEL staining. Together, these findings define a novel pathway regulated by GLI1 controlling pancreatic tumor progression and provide a new theoretical framework to help with the design and analysis of trials targeting GLI1-related pathways.


Subject(s)
Kruppel-Like Transcription Factors/antagonists & inhibitors , Pancreatic Neoplasms/pathology , Animals , Disease Progression , Humans , Mice , Mice, Knockout , Zinc Finger Protein GLI1
12.
Epigenetics Chromatin ; 6(1): 3, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23448518

ABSTRACT

BACKGROUND: Histone methyltransferase enhancer of zeste homologue 2 (EZH2) forms an obligate repressive complex with suppressor of zeste 12 and embryonic ectoderm development, which is thought, along with EZH1, to be primarily responsible for mediating Polycomb-dependent gene silencing. Polycomb-mediated repression influences gene expression across the entire gamut of biological processes, including development, differentiation and cellular proliferation. Deregulation of EZH2 expression is implicated in numerous complex human diseases. To date, most EZH2-mediated function has been primarily ascribed to a single protein product of the EZH2 locus. RESULTS: We report that the EZH2 locus undergoes alternative splicing to yield at least two structurally and functionally distinct EZH2 methyltransferases. The longest protein encoded by this locus is the conventional enzyme, which we refer to as EZH2α, whereas EZH2ß, characterized here, represents a novel isoform. We find that EZH2ß localizes to the cell nucleus, complexes with embryonic ectoderm development and suppressor of zeste 12, trimethylates histone 3 at lysine 27, and mediates silencing of target promoters. At the cell biological level, we find that increased EZH2ß induces cell proliferation, demonstrating that this protein is functional in the regulation of processes previously attributed to EZH2α. Biochemically, through the use of genome-wide expression profiling, we demonstrate that EZH2ß governs a pattern of gene repression that is often ontologically redundant from that of EZH2α, but also divergent for a wide variety of specific target genes. CONCLUSIONS: Combined, these results demonstrate that an expanded repertoire of EZH2 writers can modulate histone code instruction during histone 3 lysine 27-mediated gene silencing. These data support the notion that the regulation of EZH2-mediated gene silencing is more complex than previously anticipated and should guide the design and interpretation of future studies aimed at understanding the biochemical and biological roles of this important family of epigenomic regulators.

13.
Hum Immunol ; 74(2): 145-50, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23137876

ABSTRACT

Estimates of T regulatory cell populations in the periphery of patients with Crohn's disease are confounded by disease activity and concomitant immunotherapeutic agents known to affect T cell proliferation and survival. We performed deuterium pulse/chase experiments in patients with quiescent Crohn's disease on no immunotherapy and healthy control subjects to estimate T regulatory cell kinetics. Quantification of deuterated DNA isolated from T cell subsets over 10 days was determined by mass spectrophotometry. We demonstrate enhanced proliferation within the T regulatory cell population from patients with Crohn's disease when compared to non-T regulatory cells and T regulatory cells from healthy control subjects. We speculate that T regulatory cells isolated from the periphery of patients with Crohn's disease experience persistent antigen stimulation resulting in excess proliferative rates.


Subject(s)
Crohn Disease/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Apoptosis/immunology , Blood Glucose , Case-Control Studies , Crohn Disease/metabolism , Female , Humans , Immunologic Memory , Lymphocyte Activation/immunology , Male , Middle Aged , T-Lymphocytes, Regulatory/metabolism , Young Adult
14.
J Biol Chem ; 287(41): 34372-85, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-22896699

ABSTRACT

Inducible gene expression underlies the epigenetically inherited differentiation program of most immune cells. We report that the promoter of the FOXP3 gene possesses two distinct functional states: an "off state" mediated by the polycomb histone methyltransferase complex and a histone acetyltransferase-dependent "on state." Regulating these states is the presence of a Kruppel-like factor (KLF)-containing Polycomb response element. In the KLF10(-/-) mouse, the FOXP3 promoter is epigenetically silenced by EZH2 (Enhancer of Zeste 2)-mediated trimethylation of Histone 3 K27; thus, impaired FOXP3 induction and inappropriate adaptive T regulatory cell differentiation results in vitro and in vivo. The epigenetic transmittance of adaptive T regulatory cell deficiency is demonstrated throughout more than 40 generations of mice. These results provide insight into chromatin remodeling events key to phenotypic features of distinct T cell populations.


Subject(s)
Early Growth Response Transcription Factors/biosynthesis , Forkhead Transcription Factors/biosynthesis , Gene Silencing/physiology , Kruppel-Like Transcription Factors/biosynthesis , Polycomb-Group Proteins/metabolism , Response Elements/physiology , T-Lymphocytes, Regulatory/metabolism , p300-CBP Transcription Factors/metabolism , Animals , Chromatin Assembly and Disassembly/physiology , Early Growth Response Transcription Factors/genetics , Early Growth Response Transcription Factors/immunology , Enhancer of Zeste Homolog 2 Protein , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Male , Mice , Mice, Knockout , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/immunology , Polycomb Repressive Complex 2/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/immunology
15.
J Immunol ; 184(12): 7247-56, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20483763

ABSTRACT

The expression of pathogen recognition receptors in human FOXP3+ T regulatory cells is established, yet the function of these receptors is currently obscure. In the process of studying the function of both peripheral and lamina propria FOXP3+ lymphocytes in patients with the human inflammatory bowel disease Crohn's disease, we observed a clear deficiency in the quantity of FOXP3+ lymphocytes in patients with disease-associated polymorphisms in the pathogen recognition receptor gene NOD2. Subsequently, we determined that the NOD2 ligand, muramyl dipeptide (MDP), activates NF-kappaB in primary human FOXP3+ T cells. This activation is functionally relevant, as MDP-stimulated human FOXP3+ T cells are protected from death receptor Fas-mediated apoptosis. Importantly, apoptosis protection was not evident in MDP-stimulated FOXP3+ T cells isolated from a patient with the disease-associated polymorphism. Thus, we propose that one function of pathogen recognition receptors in human T regulatory cells is the protection against death receptor-mediated apoptosis in a Fas ligand-rich environment, such as that of the inflamed intestinal subepithelial space.


Subject(s)
Apoptosis/immunology , Crohn Disease/immunology , Forkhead Transcription Factors/immunology , Nod2 Signaling Adaptor Protein/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Apoptosis/genetics , Blotting, Western , Cell Separation , Cell Survival , Crohn Disease/genetics , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Forkhead Transcription Factors/metabolism , Genotype , Humans , Immunohistochemistry , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Male , Middle Aged , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Polymorphism, Single Nucleotide , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Transfection
16.
J Immunol ; 179(3): 1893-900, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17641056

ABSTRACT

Although functionally relevant TLRs can be expressed on human T regulatory (Treg) cells, little is known about the transcriptional control of their expression. We hypothesized that the transcription factor forkhead box P3 (FOXP3) regulates the expression of TLR family members in human Treg cells. Using primary human T cells and a reporter assay in Jurkat T cell lines, we dissected the regulation of TLR10, a TLR highly expressed in human Treg cells. We determined that TLR10 was expressed in human Treg cells through quantitative PCR, Western blotting, and flow cytometry. DNA binding of FOXP3 to a suspected cis-regulatory region in proximity to the transcription start site of TLR10 was established through EMSA and chromatin immunoprecipitation. Transcriptional control of TLR10 by FOXP3 was determined through luciferase reporter assays in Jurkat T cell lines. Relevance of FOXP3 to TLR10 gene transcription in primary T cells was established through the transfection of primary CD4(+)CD25(-)FOXP3(-) T cells with a FOXP3 expression vector, which resulted in prompt production of TLR10 mRNA. Enhanced expression of TLR10 protein in primary Treg cells was induced in a calcium-dependent fashion through TCR activation. The suspected promotional cooperation between FOXP3 and NF-AT was established in the abolition of the luciferase signal upon transfection of a mutant FOXP3 devoid of NF-AT-binding activity. These results suggest that human Treg cells express TLR10, and this expression is regulated through a cooperative complex of FOXP3 and NF-AT.


Subject(s)
Forkhead Transcription Factors/physiology , Gene Expression Regulation/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 10/biosynthesis , Toll-Like Receptor 10/genetics , Consensus Sequence/immunology , Forkhead Transcription Factors/metabolism , Humans , Jurkat Cells , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/physiology , Promoter Regions, Genetic/immunology , Protein Binding/immunology , Transcription Initiation Site , Transcriptional Activation/immunology
17.
Clin Cancer Res ; 11(18): 6641-9, 2005 Sep 15.
Article in English | MEDLINE | ID: mdl-16166443

ABSTRACT

PURPOSE: To assess the maximum tolerated dose, toxicities, pharmacokinetics, and antileukemic activity of topotecan and carboplatin in adults with recurrent or refractory acute leukemias. EXPERIMENTAL DESIGN: Patients received topotecan and carboplatin by 5-day continuous infusion at nine dose levels. Patients achieving a complete remission received up to two additional courses for consolidation. Plasma topotecan and ultrafilterable platinum were assayed on days 1 to 5. In addition, pretreatment levels of various polypeptides in leukemic cells were examined by immunoblotting to assess possible correlations with response. RESULTS: Fifty-one patients received a total of 69 courses of therapy. Dose-limiting toxicity consisted of grade 4/5 typhlitis and grade 3/4 mucositis after one course of therapy or grade 4 neutropenia and thrombocytopenia lasting >50 days when a second course was administered on day 21. Among 45 evaluable patients, there were 7 complete remissions, 2 partial remissions, 1 incomplete complete remission, and 1 reversion to chronic-phase chronic myelogenous leukemia. Topotecan steady-state plasma concentrations increased with dose. No accumulation of topotecan or ultrafilterable platinum occurred between days 1 and 5 of therapy. Leukemic cell levels of topoisomerase I, checkpoint kinase 1, checkpoint kinase 2, and Mcl-1 correlated with proliferating cell nuclear antigen but not with response. In contrast, low Bcl-2 expression correlated with response (P = 0.014, Mann-Whitney U test). CONCLUSIONS: The maximum tolerated dose was 1.6 mg/m(2)/d topotecan plus 150 mg/m(2)/d carboplatin. The complete remission rate in a heavily pretreated population was 16% (33% at the highest three dose levels). Responses seem to correlate with low pretreatment blast cell Bcl-2 expression.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia/drug therapy , Acute Disease , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Carboplatin/administration & dosage , Carboplatin/adverse effects , Carboplatin/pharmacokinetics , Cell Cycle Proteins/metabolism , Combined Modality Therapy , DNA Topoisomerases, Type I/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Female , HL-60 Cells , Hematopoietic Stem Cell Transplantation , Humans , Immunoblotting , Infusions, Intravenous , Leukemia/metabolism , Male , Middle Aged , Neoplasm Recurrence, Local , Proliferating Cell Nuclear Antigen/metabolism , Topotecan/administration & dosage , Topotecan/adverse effects , Topotecan/pharmacokinetics , Treatment Outcome
18.
Clin Cancer Res ; 10(20): 6807-20, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15501957

ABSTRACT

PURPOSE: According to some studies, susceptibility of cells to anticancer drug-induced apoptosis is markedly inhibited by targeted deletion of genes encoding apoptotic protease activating factor 1 (Apaf-1) or certain caspases. Information about levels of these polypeptides in common cancer cell types and any possible correlation with drug sensitivity in the absence of gene deletion is currently fragmentary. EXPERIMENTAL DESIGN: Immunoblotting was used to estimate levels of Apaf-1 as well as procaspase-2, -3, -6, -7, -8, and -9 in the 60-cell-line panel used for drug screening by the National Cancer Institute. Sensitivity of the same lines to >80,000 compounds was determined with 48-hour sulforhodamine B binding assays. Additional 6-day assays were performed for selected agents. RESULTS: Levels of Apaf-1 and procaspases varied widely. Apaf-1 and procaspase-9, which are implicated in caspase activation after treatment of cells with various anticancer drugs, were detectable in all of the cell lines, with levels of Apaf-1 ranging from approximately 1 x 10(5) to 2 x 10(6) molecules per cell and procaspase-9 from approximately 5 x 10(3) to approximately 1.6 x 10(5) molecules per cell. Procaspase-8 levels ranged from 1.7 x 10(5) to 8 x 10(6) molecules per cell. Procaspase-3, a major effector caspase, varied from undetectable to approximately 1.6 x 10(6) molecules per cell. Correlations between levels of these polypeptides and sensitivity to any of a variety of experimental or conventional antineoplastic agents in either 2-day or 6-day cytotoxicity assays were weak at best. CONCLUSIONS: With the exception of caspase-3, all of the components of the core cell-death machinery are expressed in all of the cell lines examined. Despite variations in expression, levels of any one component are not a major determinant of drug sensitivity in these cells in vitro.


Subject(s)
Apoptosis/drug effects , Caspases/biosynthesis , Caspases/genetics , Neoplasms/genetics , Neoplasms/pathology , Proteins/genetics , Tumor Cells, Cultured , Apoptotic Protease-Activating Factor 1 , Drug Screening Assays, Antitumor , Humans , Immunoblotting , Proteins/analysis
19.
Mol Pharmacol ; 64(6): 1434-43, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14645674

ABSTRACT

The mechanism of action of fenretinide, a synthetic retinoid currently undergoing testing as a chemopreventive and chemotherapeutic agent, is incompletely understood. In the present study, fenretinide caused apoptotic changes, including DNA fragmentation and cleavage of caspase substrates, in six low-passage ovarian cancer cell lines. However, the caspase activation pathway used by this agent varied. Transient transfection of cDNA-encoding cytokine response modifier A (CrmA), a caspase-8 inhibitor, diminished fenretinide-induced death in OV177 cells. Likewise, IETD(OMe)-fluoromethylketone (fmk) inhibited fenretinide-induced apoptosis by >80% in OV177 or OV266 cells and by approximately 50% in OV17, OV167, or OV207 cells. Further analysis demonstrated that inhibition of Fas ligand, tumor necrosis factor-alpha, or TRAIL signaling with blocking reagents did not affect fenretinide-induced apoptosis, raising the possibility that fenretinide activates caspase-8 in a death receptor-independent manner. In contrast, CrmA transfection or IETD(OMe)-fmk treatment did not inhibit fenretinide-induced apoptosis in OV202 cells. These divergent behaviors did not correlate with increased levels of procaspase-10, which is relatively resistant to CrmA and IETD(OMe)-fmk, nor with the expression of procaspase-8 and -9, apoptotic protease activating factor-1, or cellular FLICE-like inhibitory protein. Similarly, fenretinide treatment increased ceramide levels equally in cells that do (OV177) and do not (OV202) rely on caspase-8 to initiate apoptosis. These results indicate that synthetic retinoids can use caspase-8 as an initiating caspase, but they also indicate unexpected heterogeneity in caspase activation pathways among closely related cell lines.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Fenretinide/pharmacology , Neoplasms, Glandular and Epithelial/enzymology , Ovarian Neoplasms/enzymology , Apoptosis/physiology , Caspase 8 , Caspase 9 , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Female , Humans
20.
Blood ; 99(2): 664-71, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11781252

ABSTRACT

The adenosine triphosphate binding-site-directed agent STI571 and the tyrphostin adaphostin are undergoing evaluation as bcr/abl kinase inhibitors. The current study compared the effects of these agents on the survival of K562 cells, bcr/abl-transduced FDC-P1 cells, and myeloid progenitors from patients with chronic myelogenous leukemia (CML) compared with healthy donors. Treatment of K562 cells with 10 microM adaphostin resulted in decreased p210(bcr/abl) polypeptide levels in the first 6 hours, followed by caspase activation and accumulation of apoptotic cells in less than 12 hours. By 24 hours, 90% of the cells were apoptotic and unable to form colonies. In contrast, 20 microM STI571 caused rapid inhibition of bcr/abl autophosphorylation without p210(bcr/abl) degradation. Although this was followed by the inhibition of Stat5 phosphorylation and the down-regulation of Bcl-x(L) and Mcl-1, only 7% +/- 3% and 25% +/- 9% of cells were apoptotic at 16 and 24 hours, respectively. Instead, the cytotoxic effects of STI571 became more pronounced with prolonged exposure, with IC90 values greater than 20 microM and 1.0 +/- 0.6 microM after 24 and 48 hours, respectively. Consistent with these results, 24-hour adaphostin exposure inhibited CML granulocyte colony-forming units (CFU-G) (median IC50, 12 microM) but not normal CFU-G (median IC50, greater than 20 microM), whereas 24-hour STI571 treatment had no effect on CML or normal CFU-G. Additional experiments revealed that STI571-resistant K562 cells remained sensitive to adaphostin. Moreover, the combination of STI571 + adaphostin induced more cytotoxicity in K562 cells and in CML CFU-G than either agent alone did. Collectively, these results identify adaphostin as a mechanistically distinct CML-selective agent that retains activity in STI571-resistant cell lines.


Subject(s)
Adamantane/pharmacology , Enzyme Inhibitors/pharmacology , Fusion Proteins, bcr-abl/antagonists & inhibitors , Hydroquinones/pharmacology , K562 Cells/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Viral Proteins , Adamantane/analogs & derivatives , Apoptosis/drug effects , Benzamides , Caspase 9 , Caspases/genetics , Caspases/metabolism , Drug Synergism , Enzyme Induction , Fusion Proteins, bcr-abl/biosynthesis , Fusion Proteins, bcr-abl/genetics , Humans , Imatinib Mesylate , K562 Cells/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Phosphorylation/drug effects , Protein Biosynthesis , Protein Processing, Post-Translational/drug effects , Proteins/genetics , Recombinant Fusion Proteins/physiology , Serpins/genetics , Serpins/metabolism , Transfection , Tumor Cells, Cultured/drug effects , Tumor Stem Cell Assay , X-Linked Inhibitor of Apoptosis Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...