Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
bioRxiv ; 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38405748

ABSTRACT

Inflammatory Bowel Disease ( IBD ) is a chronic and often debilitating autoinflammatory condition, with an increasing incidence in children. Standard-of-care therapies lead to sustained transmural healing and clinical remission in fewer than one-third of patients. For children, TNFα inhibition remains the only FDA-approved biologic therapy, providing an even greater urgency to understanding mechanisms of response. Genome-wide association studies ( GWAS ) have identified 418 independent genetic risk loci contributing to IBD, yet the majority are noncoding and their mechanisms of action are difficult to decipher. If causal, they likely alter transcription factor ( TF ) binding and downstream gene expression in particular cell types and contexts. To bridge this knowledge gap, we built a novel resource: multiome-seq (tandem single-nuclei ( sn )RNA-seq and chromatin accessibility ( snATAC )-seq) of intestinal tissue from pediatric IBD patients, where anti-TNF response was defined by endoscopic healing. From the snATAC-seq data, we generated a first-time atlas of chromatin accessibility (putative regulatory elements) for diverse intestinal cell types in the context of IBD. For cell types/contexts mediating genetic risk, we reasoned that accessible chromatin will co-localize with genetic disease risk loci. We systematically tested for significant co-localization of our chromatin accessibility maps and risk variants for 758 GWAS traits. Globally, genetic risk variants for IBD, autoimmune and inflammatory diseases are enriched in accessible chromatin of immune populations, while other traits (e.g., colorectal cancer, metabolic) are enriched in epithelial and stromal populations. This resource opens new avenues to uncover the complex molecular and cellular mechanisms mediating genetic disease risk.

2.
bioRxiv ; 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38352573

ABSTRACT

Background and Aims: We previously identified small molecules predicted to reverse an ileal gene signature for future Crohn's Disease (CD) strictures. Here we used a new human intestinal organoid (HIO) model system containing macrophages to test a lead candidate, eicosatetraynoic acid (ETYA). Methods: Induced pluripotent stem cell lines (iPSC) were derived from CD patients and differentiated into macrophages and HIOs. Macrophages and macrophage:HIO co-cultures were exposed to lipopolysaccharide (LPS) with and without ETYA pre-treatment. Cytospin and flow cytometry characterized macrophage morphology and activation markers, and RNA sequencing defined the global pattern of macrophage gene expression. TaqMan Low Density Array, Luminex multiplex assay, immunohistologic staining, and sirius red polarized light microscopy were performed to measure macrophage cytokine production and HIO pro-fibrotic gene expression and collagen content. Results: iPSC-derived macrophages exhibited morphology similar to primary macrophages and expressed inflammatory macrophage cell surface markers including CD64 and CD68. LPS-stimulated macrophages expressed a global pattern of gene expression enriched in CD ileal inflammatory macrophages and matrisome secreted products, and produced cytokines and chemokines including CCL2, IL1B, and OSM implicated in refractory disease. ETYA suppressed CD64 abundance and pro-fibrotic gene expression pathways in LPS stimulated macrophages. Co-culture of LPS-primed macrophages with HIO led to up-regulation of fibroblast activation genes including ACTA2 and COL1A1 , and an increase in HIO collagen content. ETYA pre-treatment prevented pro-fibrotic effects of LPS-primed macrophages. Conclusions: ETYA inhibits pro-fibrotic effects of LPS-primed macrophages upon co-cultured HIO. This model may be used in future untargeted screens for small molecules to treat refractory CD.

3.
Inflamm Bowel Dis ; 28(7): 988-1003, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35259271

ABSTRACT

BACKGROUND: Perturbagen analysis of Crohn's disease (CD) ileal gene expression data identified small molecules including eicosatetraynoic acid (ETYA), which may exert an antifibrotic effect. We developed a patient-specific human intestinal organoid (HIO) model system to test small molecule regulation of mitochondrial and wound-healing functions implicated in stricturing behavior. METHODS: HIOs were made from CD induced pluripotent stem cells with and without a loss-of-function haplotype in the DUOX2 gene implicated in ileal homeostasis and characterized under basal conditions and following exposure to butyrate and ETYA using RNA sequencing, flow cytometry, and immunofluorescent and polarized light microscopy. Mitochondrial activity was measured using high-resolution respirometry and tissue stiffness using atomic force microscopy. RESULTS: HIOs expressed core mitochondrial and extracellular matrix (ECM) genes and enriched biologic functions implicated in CD ileal strictures; ECM gene expression was suppressed by both butyrate and ETYA, with butyrate also suppressing genes regulating epithelial proliferation. Consistent with this, butyrate, but not ETYA, exerted a profound effect on HIO epithelial mitochondrial function, reactive oxygen species production, and cellular abundance. Butyrate and ETYA suppressed HIO expression of alpha smooth muscle actin expressed by myofibroblasts, type I collagen, and collagen protein abundance. HIOs exhibited tissue stiffness comparable to normal human ileum; this was reduced by chronic ETYA exposure in HIOs carrying the DUOX2 loss-of-function haplotype. CONCLUSIONS: ETYA regulates ECM genes implicated in strictures and suppresses collagen content and tissue stiffness in an HIO model. HIOs provide a platform to test personalized therapeutics, including small molecules prioritized by perturbagen analysis.


A subset of pediatric Crohn's disease patients develop intestinal strictures requiring surgery. The microbial metabolite butyrate and eicosatetraynoic acid regulate pathways implicated in stricture formation in a human intestinal organoid model system, which may be used to test new therapies.


Subject(s)
Crohn Disease , Butyrates/metabolism , Butyrates/pharmacology , Collagen/metabolism , Constriction, Pathologic/metabolism , Crohn Disease/genetics , Dual Oxidases/metabolism , Extracellular Matrix/metabolism , Humans , Intestinal Mucosa/metabolism , Mitochondria/metabolism , Organoids/metabolism
4.
Sci Rep ; 9(1): 9168, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31235766

ABSTRACT

Neutrophil dysfunction and GM-CSF auto-antibodies are observed in pediatric and adult patients with Crohn's disease (CD). We associated damaging coding variants with low GM-CSF induced STAT5 stimulation index (GMSI) in pediatric CD patients and implicated variation of neutrophil GM-CSF signaling in cell function and disease complications. Because many CD patients with low GMSI do not carry damaging coding mutations, we sought to test the hypothesis that non-coding variants contribute to this phenotype. We enrolled, performed whole genome sequencing, and measured the GMSI in 77 CD and ulcerative colitis (UC) patients (24 low and 53 normal GMSI). We identified 4 non-coding variants (rs3808851, rs10974787, rs10974788 and rs10974789) in RCL1 significantly associated with variation of GMSI level (p < 0.011). They were validated in two independent cohorts with: RNAseq data (n = 50) and blood eQTL dataset (n = 31,684). These variants are in LD and affect expression of JAK2 (p 0.005 to 0.013), RCL1 (p 8.17E-13 to 2.98E-11) and AK3 (p 2.00E-68 to 3.03E-55) genes. Additionally, they influence proteins involved in differentiation of gut epithelium, inflammation, and immune system regulation. In summary, our study outlines the contribution of non-coding variants in neutrophil GM-CSF signaling and the potential importance of RCL1 and AK3 in CD pathogenesis.


Subject(s)
Crohn Disease/genetics , Crohn Disease/pathology , Genetic Variation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Neutrophils/pathology , Signal Transduction/genetics , Child , Female , Humans , Male
5.
J Pediatr ; 210: 220-225, 2019 07.
Article in English | MEDLINE | ID: mdl-31027832

ABSTRACT

A 4-year-old boy presented with perianal abscess and granulomatous colitis, which led the diagnosis of Crohn's disease. He became refractory to all available therapies and required colectomy. Targeted sequencing revealed a deleterious variant in NCF4, causing severe neutrophil dysfunction. He underwent hematopoietic stem cell transplantation (HSCT) with an excellent outcome.


Subject(s)
Crohn Disease/genetics , Crohn Disease/therapy , Hematopoietic Stem Cell Transplantation , Mutation , NADPH Oxidases/genetics , Child, Preschool , Crohn Disease/diagnosis , Humans , Male
6.
Nat Commun ; 10(1): 38, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30604764

ABSTRACT

Molecular mechanisms driving disease course and response to therapy in ulcerative colitis (UC) are not well understood. Here, we use RNAseq to define pre-treatment rectal gene expression, and fecal microbiota profiles, in 206 pediatric UC patients receiving standardised therapy. We validate our key findings in adult and paediatric UC cohorts of 408 participants. We observe a marked suppression of mitochondrial genes and function across cohorts in active UC, and that increasing disease severity is notable for enrichment of adenoma/adenocarcinoma and innate immune genes. A subset of severity genes improves prediction of corticosteroid-induced remission in the discovery cohort; this gene signature is also associated with response to anti-TNFα and anti-α4ß7 integrin in adults. The severity and therapeutic response gene signatures were in turn associated with shifts in microbes previously implicated in mucosal homeostasis. Our data provide insights into UC pathogenesis, and may prioritise future therapies for nonresponders to current approaches.


Subject(s)
Colitis, Ulcerative/genetics , Genes, Mitochondrial/genetics , Intestinal Mucosa/metabolism , Mitochondrial Diseases/genetics , Transcriptome/genetics , Adolescent , Adult , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Child , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Feces/microbiology , Female , Gene Expression Profiling , Glucocorticoids/therapeutic use , Humans , Integrins/antagonists & inhibitors , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mesalamine/therapeutic use , Microbiota , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/microbiology , Mitochondrial Diseases/pathology , Precision Medicine/methods , Prospective Studies , Rectum/metabolism , Rectum/microbiology , Rectum/pathology , Remission Induction/methods , Sequence Analysis, RNA , Severity of Illness Index , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors
7.
Inflamm Bowel Dis ; 25(3): 547-560, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30124884

ABSTRACT

BACKGROUND: Granulocyte-macrophage colony-stimulating factor auto-antibodies (GMAbs) suppress neutrophil-extrinsic GM-CSF signaling and increase risk for stricturing behavior in Crohn's disease (CD). We aimed to define clinical, genomic, and functional associations with neutrophil-intrinsic GM-CSF signaling. METHODS: Missense mutations in CSF2RA, CSF2RB, JAK2, STAT5A, and STAT5B were identified using whole-exome sequencing in 543 pediatric inflammatory bowel disease (IBD) patients. Neutrophil-intrinsic GM-CSF signaling was defined using the GM-CSF-induced STAT5 stimulation index (GMSI) in 180 pediatric IBD patients and 26 non-IBD controls. Reduced GM-CSF signaling (GMSI-Lo) was defined as the 20th percentile within the control group. Variation in neutrophil phospho-protein abundance, bacterial killing, and the global pattern of gene expression with the GMSI was determined. RESULTS: We validated 18 potentially damaging missense mutations in CSF2RA and CSF2RB. CSF2RA A17G carriage increased from 10% in those with intact neutrophil GMSI to 32% in those with low GMSI (P = 0.02). The frequency of reduced Staphylococcus aureus killing increased from 17% in those with intact neutrophil GMSI to 35% in GMSI-Lo neutrophils (P = 0.043). Crohn's disease neutrophils with low GMSI exhibited specific alterations in phospho-protein networks and genes regulating cytokine production, wound healing, and cell survival and proliferation. Stricturing behavior increased from 7% in patients with both low GMAb and intact GMSI to 64% in patients with both elevated GMAb and low GMSI (P < 0.0001). CONCLUSIONS: Low/normal neutrophil-intrinsic GM-CSF signaling is associated with CSF2RA missense mutations, alterations in gene expression networks, and higher rates of disease complications in pediatric CD.


Subject(s)
Crohn Disease/pathology , Cytokine Receptor Common beta Subunit/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Mutation, Missense , Neutrophils/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Transcriptome , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Crohn Disease/genetics , Crohn Disease/metabolism , Female , Follow-Up Studies , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Infant , Male , Neutrophils/metabolism , Prognosis , Young Adult
8.
Gastroenterology ; 154(8): 2097-2110, 2018 06.
Article in English | MEDLINE | ID: mdl-29454792

ABSTRACT

BACKGROUND & AIMS: Individuals with monogenic disorders of phagocyte function develop chronic colitis that resembles Crohn's disease (CD). We tested for associations between mutations in genes encoding reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, neutrophil function, and phenotypes of CD in pediatric patients. METHODS: We performed whole-exome sequence analysis to identify mutations in genes encoding NADPH oxidases (such as CYBA, CYBB, NCF1, NCF2, NCF4, RAC1, and RAC2) using DNA from 543 pediatric patients with inflammatory bowel diseases. Blood samples were collected from an additional 129 pediatric patients with CD and 26 children without IBD (controls); we performed assays for neutrophil activation, reactive oxygen species (ROS) production, and bacteria uptake and killing. Whole-exome sequence analysis was performed using DNA from 46 of the children with CD to examine associations with NADPH gene mutations; RNA sequence analyses were performed using blood cells from 46 children with CD to test for variations in neutrophil gene expression associated with ROS production. RESULTS: We identified 26 missense mutations in CYBA, CYBB, NCF1, NCF2, and NCF4. Patients with CD who carried mutations in these genes were 3-fold more likely to have perianal disease (P = .0008) and stricturing complications (P = .002) than children with CD without these mutations. Among patients with CD with none of these mutations, 9% had undergone abdominal surgery; among patients with mutations in these NADPH oxidase genes, 31% had undergone abdominal surgery (P = .0004). A higher proportion of neutrophils from children with CD had low ROS production (47%) than from controls (15%) among the 129 patients tested for ROS (P = .002). Minor alleles of the NADPH genes were detected in 7% of children with CD whose neutrophils produced normal levels of ROS vs 38% of children whose neutrophils produced low levels of ROS (P = .009). Neutrophils that produced low levels of ROS had specific alterations in genes that regulate glucose metabolism and antimicrobial responses. CONCLUSIONS: We identified missense mutations in genes that encode NADPH oxidases in children with CD; these were associated with a more aggressive disease course and reduced ROS production by neutrophils from the patients.


Subject(s)
Crohn Disease/genetics , NADPH Oxidases/genetics , Neutrophils/metabolism , Reactive Oxygen Species/metabolism , Adolescent , Alleles , Child , Child, Preschool , Cohort Studies , Crohn Disease/blood , Crohn Disease/metabolism , Down-Regulation , Female , Gene Expression Profiling , Glucose/metabolism , Humans , Infant , Male , Mutation, Missense , Phenotype , Sequence Analysis, RNA , Up-Regulation , Exome Sequencing
9.
Inflamm Bowel Dis ; 20(6): 1037-48, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24788216

ABSTRACT

BACKGROUND: Neutrophil expression of the Fcγ receptor I (CD64) is upregulated in adult patients with clinically active inflammatory bowel disease (IBD). We tested the relationship of CD64 with mucosal inflammation and clinical relapse in pediatric Crohn's disease (CD). METHODS: In a cohort of 208 newly diagnosed CD and 43 non-IBD controls, ileal expression of FcγRI/S100A9 was determined by RNA sequencing from biopsies obtained at ileocolonoscopy. In a second cohort, we tested for the peripheral blood polymorphonuclear neutrophil (PMN) CD64 index from 26 newly diagnosed CD, 30 non-IBD controls, and 83 children with established CD. RESULTS: Ileal FcγRIA mRNA expression was significantly elevated in CD at diagnosis compared with non-IBD controls (P < 0.001), and correlated with ileal S100A9 (calprotectin) expression (r = 0.83, P < 0.001). The median (range) PMN CD64 index for newly diagnosed CD was 2.3 (0.74-9.3) compared with 0.76 (0.39-1.2) for non-IBD controls (P < 0.001) with 96% sensitivity and 90% specificity at the cut point of 1.0. The PMN CD64 index significantly correlated with mucosal injury as measured by the simple endoscopic score for CD (r = 0.62, P < 0.001). Patients with CD in clinical remission receiving maintenance therapy with a PMN CD64 index <1.0 had a sustained remission rate of 95% over the following 12 months compared with 56% in those with a PMN CD64 index >1.0 (P < 0.01). CONCLUSIONS: An elevated PMN CD64 index is associated with both mucosal inflammation and an increased risk for clinical relapse in pediatric CD. The PMN CD64 index is a reliable marker for sustained remission in patients with CD receiving maintenance therapy.


Subject(s)
Crohn Disease/diagnosis , Intestinal Mucosa/metabolism , Neutrophils/metabolism , Receptors, IgG/metabolism , Adolescent , Biomarkers/metabolism , Calgranulin B/genetics , Calgranulin B/metabolism , Child , Child, Preschool , Cohort Studies , Crohn Disease/epidemiology , Crohn Disease/metabolism , Female , Humans , Ileum/immunology , Ileum/metabolism , Intestinal Mucosa/immunology , Male , Neutrophils/immunology , Receptors, IgG/genetics , Rectum/immunology , Rectum/metabolism , Recurrence , Remission Induction , Risk Factors , Sensitivity and Specificity
10.
Inflamm Bowel Dis ; 19(3): 512-25, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23429443

ABSTRACT

BACKGROUND: Intestinal epithelial cell (IEC) STAT3 is required for wound healing following acute dextran sodium sulfate (DSS) injury. We hypothesized that loss of IEC STAT3 would promote the development of chronic colitis following acute DSS injury. METHODS: Colitis was induced in IEC-specific STAT3-deficient mice (STAT3)[INCREMENT]IEC and littermate controls (STAT3 Flx/Flx) with 4% DSS for 7 days, followed by water consumption for 21 days. Epithelial and immune mediators and severity of colitis were determined. RESULTS: Survival, colon length, and histologic injury were significantly worse at day 28 in STAT3[INCREMENT]IEC mice. IEC proliferation and apoptosis did not vary by genotype at day 14 or day 28. The colonic lamina propria frequency of pSTAT3* cells was increased at day 28 and correlated with histologic injury in STAT3 [INCREMENT]IEC mice. The frequency of colonic F480* pSTAT3* macrophages and CD3* pSTAT3* T lymphocytes were increased in STAT3[INCREMENT]IEC mice as compared with STAT3 Flx/Flx controls. In STAT3[INCREMENT]IEC mice, colonic expression of STAT3 target genes Reg3ß and Reg3γ, which mediate epithelial restitution, were significantly decreased, whereas expression of interleukin (IL)-17a, IFNγ, CXCL2, CXCL10, and CCL2 were significantly increased and correlated with the increase in histologic severity at day 28(P < 0.05). IL-17a expression also correlated with the increased lamina propria frequency of CD3* pSTAT3* T lymphocytes. CONCLUSIONS: Loss of intestinal epithelial STAT3 leads to more severe chronic inflammation following acute injury, which is not accounted for by a sustained defect in epithelial proliferation or apoptosis 7 or 21 days after 1 cycle of DSS but rather defective REG3 expression and expansion of pSTAT3* lymphocytes and IL-17A expression.


Subject(s)
Colitis/metabolism , Intestinal Mucosa/metabolism , STAT3 Transcription Factor/deficiency , Acute Disease , Animals , Biomarkers/metabolism , Chronic Disease , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Cytokines/metabolism , Dextran Sulfate , Fluorescent Antibody Technique , Immunohistochemistry , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Macrophages/metabolism , Mice , Mice, Knockout , Pancreatitis-Associated Proteins , Proteins/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Severity of Illness Index , T-Lymphocytes/metabolism
11.
J Pediatr Gastroenterol Nutr ; 55(1): 32-43, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22197944

ABSTRACT

OBJECTIVES: Genotypic variation in signal transducer and activator of transcription 3 (STAT3) increases risk for inflammatory bowel disease (IBD), and STAT3-dependent inflammatory networks are induced in the colon in these patients. We hypothesized that STAT3 "A" risk allele carriage would be associated with increased cellular STAT3 activation and colon leukocyte recruitment. METHODS: Colonic expression of genes regulating STAT3 signaling and leukocyte recruitment and function was measured in pediatric patients with Crohn disease (CD) stratified by STAT3 genotype. The frequency of colonic pSTAT3* and CXCR2* neutrophils was determined using immunohistochemistry. STAT3 tyrosine phosphorylation (pSTAT3) was measured in circulating leukocytes by flow cytometry, and mechanisms regulating STAT3 activation were tested in IBD Epstein-Barr virus (EBV)-transformed lymphocytes (EBL). RESULTS: Colonic expression of interleukin 6 (IL-6), the STAT3 target gene SOCS3, the neutrophil chemoattractants IL-8, CXCL1, and CXCL3, and the neutrophil products S100A8, S100A9, and S100A12 were increased in patients carrying the STAT3 "A" risk allele. The frequency of neutrophils expressing the cognate receptor for IL-8, CXCR2, was increased in colonic biopsies from patients carrying the risk allele, and the frequency of pSTAT3* or CXCR2* neutrophils correlated with histologic severity. The frequency of CD4 lymphocytes and granulocytes expressing pSTAT3 was increased in patients carrying the STAT3 "A" risk allele. EBLs from patients carrying the STAT3 "A" risk allele exhibited increased basal and IL-6-stimulated STAT3 tyrosine phosphorylation, increased transcription of STAT3 and SOCS3 after IL-6 stimulation, and increased membrane localization of the IL-6 receptor, GP130, and Janus-associated kinase 2. CONCLUSIONS: The STAT3 "A" risk allele is associated with increased cellular STAT3 activation and upregulation of pathways that promote recruitment of CXCR2* neutrophils to the gut.


Subject(s)
Colon/metabolism , Crohn Disease/genetics , Crohn Disease/metabolism , Neutrophils/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction/genetics , Adolescent , Alleles , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes , Calgranulin A/metabolism , Calgranulin B/metabolism , Cells, Cultured , Chemokine CXCL1/metabolism , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Child , Colon/immunology , Colon/pathology , Crohn Disease/pathology , Female , Gene Expression , Genetic Variation , Genotype , Humans , Interleukin-1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Janus Kinase 2/metabolism , Lymphocyte Count , Male , Phosphorylation , Polymorphism, Single Nucleotide , Receptors, Interleukin-8B/metabolism , S100 Proteins/metabolism , S100A12 Protein , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Tyrosine/metabolism , Up-Regulation
12.
Inflamm Bowel Dis ; 17(12): 2443-55, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21381154

ABSTRACT

BACKGROUND: Ileal involvement in Crohn's disease (CD) is associated with NOD2 mutations and granulocyte-macrophage colony stimulating factor autoantibodies (GM-CSF Ab), and GM-CSF blockade promotes ileitis in Nod2/Card15-deficient (C15KO) mice. RALDH2-expressing dendritic cells (DC) and IL-4 promote CCR9 imprinting and small bowel homing of T lymphocytes, in conjunction with CCL25 expression by ileal epithelial cells (IEC). We hypothesized that GM-CSF neutralization promotes ileal disease by modulating expression of CCL25 by IEC and CCR9 by T lymphocytes via Nod2-dependent and independent pathways. METHODS: CCL25 and CCR9 expression were determined in pediatric CD patients stratified by GM-CSF Ab. Ileitis was induced in C15KO mice via GM-CSF Ab administration followed by nonsteroidal antiinflammatory drug (NSAID) exposure, and expression of CCL25, CCR9, FOXP3, intracellular cytokines, and RALDH2 was determined in IEC and immune cell populations. RESULTS: The frequency of CCL25(+) IEC and CCR9(+) T lymphocytes was increased in CD patients with elevated GM-CSF Ab. In the murine model, GM-CSF blockade alone induced IEC CCL25 expression, and reduced the frequency of mesenteric lymph node (MLN) CD4(+) FOXP3(+) cells, while Card15 deficiency alone enhanced MLN DC RALDH2 expression. Both GM-CSF neutralization and Card15 deficiency were required for downregulation of MLN DC IL-10 expression; under these conditions NSAID exposure led to an expansion of IL-4(+) and IL-17(+) CCR9(+) lymphocytes in the ileum. CONCLUSIONS: GM-CSF prevents ileal expansion of CCR9(+) lymphocytes via Nod2-dependent and independent pathways. CCR9 blockade may be beneficial in CD patients with elevated GM-CSF Ab.


Subject(s)
Crohn Disease/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Ileitis/pathology , Lymphocytes/pathology , Nod2 Signaling Adaptor Protein/physiology , Receptors, CCR/metabolism , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Chemokines, CC/genetics , Chemokines, CC/metabolism , Child , Crohn Disease/immunology , Crohn Disease/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Ileitis/immunology , Ileitis/metabolism , Immunoenzyme Techniques , Interleukin-10/genetics , Interleukin-10/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Mice, Knockout , Mutation/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, CCR/genetics
13.
Gut ; 59(8): 1066-78, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20584783

ABSTRACT

BACKGROUND: Administration of granulocyte-macrophage colony stimulating factor (GM-CSF) relieves symptoms in Crohn's disease (CD). It has been reported that reduced GM-CSF bioactivity is associated with more aggressive ileal behaviour and that GM-CSF-null mice exhibit ileal barrier dysfunction and develop a transmural ileitis following exposure to non-steroidal anti-inflammatory drugs (NSAIDs). STAT5 signalling is central to GM-CSF action. It was therefore hypothesised that GM-CSF signalling in non-haematopoietic cells is required for ileal homeostasis. METHODS: Bone marrow (BM) chimeras were generated by reconstituting irradiated GM-CSF receptor (gm-csfr) beta chain or GM-CSF (gm-csf) deficient mice with wild type BM (WTBM-->GMRKO and WTBM-->GMKO). Intestinal barrier function and the response to NSAID-induced ileal injury were examined. Expression of gm-csf, gm-csfr or stat5 in Caco-2 and HT-29 intestinal epithelial cell (IEC) lines was knocked down and the effect of GM-CSF signalling on IEC survival and proliferation was determined. RESULTS: Elevated levels of GM-CSF autoantibodies in ileal CD were found to be associated with dysregulation of IEC survival and proliferation. GM-CSF receptor-deficient mice and WTBM-->GMRKO chimeras exhibited ileal hyperpermeability. NSAID exposure induced a transmural ileitis in GM-CSF receptor-deficient mice and WTBM-->GMRKO chimeras. Transplantation of wild type BM into GM-CSF-deficient mice prevented NSAID ileal injury and restored ileal barrier function. Ileal crypt IEC proliferation was reduced in WTBM-->GMRKO chimeras, while STAT5 activation in ileal IEC following NSAID exposure was abrogated in WTBM-->GMRKO chimeras. Following knock down of gm-csf, gm-csfr alpha or beta chain or stat5a/b expression in Caco-2 cells, basal proliferation was suppressed. GM-CSF normalised proliferation of Caco-2 cells exposed to NSAID, which was blocked by stat5a/b RNA interference. CONCLUSIONS: Loss of GM-CSF signalling in non-haematopoietic cells increases NSAID ileal injury; furthermore, GM-CSF signalling in non-haematopoietic cells regulates ileal epithelial homeostasis via the STAT5 pathway. The therapeutic use of GM-CSF may therefore be beneficial in chronic ileitis associated with CD.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Ileitis/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal , Bone Marrow Transplantation , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Disease Models, Animal , Disease Susceptibility , Epithelial Cells/pathology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/deficiency , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Ileitis/chemically induced , Ileitis/immunology , Intestinal Mucosa/pathology , Male , Mice , Mice, Knockout , Recombinant Proteins/pharmacology , STAT5 Transcription Factor/metabolism , Signal Transduction/physiology
14.
Inflamm Bowel Dis ; 16(5): 856-69, 2010 May.
Article in English | MEDLINE | ID: mdl-19924809

ABSTRACT

BACKGROUND: Systemic exposure to lipopolysaccharide (LPS) has been linked to clinical disease activity in adults with inflammatory bowel disease (IBD). We hypothesized that markers of LPS exposure and the acute phase response (APR) would be increased in pediatric IBD patients with growth failure, and that LPS signaling would be required for induction of the APR in murine colitis. METHODS: Serum markers of LPS exposure, endotoxin core IgA antibody (EndoCAb), and the APR, LPS binding protein (LBP) were quantified in pediatric IBD patients and controls. LBP and cytokine production were determined after administration of trinitrobenzene sulfonic acid (TNBS) enemas to mice with genetic deletion of Toll-Like receptor 4 (TLR4), and wildtype (WT) controls. RESULTS: Serum EndoCAb and LBP were significantly elevated in patients with Crohn's disease (CD), compared to disease controls with ulcerative colitis (UC) and healthy controls (P < 0.001). This was independent of disease activity or location. CD patients with elevated serum EndoCAb and LBP exhibited linear growth failure which persisted during therapy. Serum LBP increased in WT mice following TNBS administration, in conjunction with increased serum TNF-alpha, IL-6, and IL-10, and expansion of regulatory T-cell numbers. Both the APR and expansion of foxp3+ T cells were abrogated in TLR4-deficient mice, in conjunction with a reduction in acute weight loss. CONCLUSIONS: LPS exposure and a persistent APR are associated with growth failure in pediatric CD. LPS signaling is required for the APR in murine colitis. Therapies targeting this pathway may benefit the subset of patients with refractory growth failure.


Subject(s)
Acute-Phase Reaction/etiology , Colitis, Ulcerative/complications , Crohn Disease/complications , Growth Disorders/etiology , Lipopolysaccharides/toxicity , Acute-Phase Proteins/metabolism , Acute-Phase Reaction/pathology , Adolescent , Adult , Animals , Biomarkers/metabolism , Carrier Proteins/metabolism , Case-Control Studies , Child , Child, Preschool , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colon/drug effects , Colon/metabolism , Crohn Disease/drug therapy , Cytokines/metabolism , Enema , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Growth Disorders/pathology , Humans , Infant , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Toll-Like Receptor 4/physiology , Trinitrobenzenesulfonic Acid/pharmacology , Young Adult
15.
Gastroenterology ; 136(4): 1261-71, e1-3, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19230854

ABSTRACT

BACKGROUND & AIMS: Genetic variations that affect innate immunity increase risk of ileal Crohn's disease (CD). However, the penetrance of susceptibility genes, including NOD2, is low, suggesting additional risk factors. Neutralizing autoantibodies (Ab) against granulocyte-macrophage colony-stimulating factor (GM-CSF Ab) reduce neutrophil antimicrobial function in patients with primary alveolar proteinosis (PAP). We investigated whether GM-CSF Ab regulates neutrophil function in CD. METHODS: Serum samples from 354 adult and pediatric patients with inflammatory bowel disease (IBD) were analyzed for GM-CSF Ab and IBD markers. Levels of GM-CSF Ab were compared with patients' CD features and neutrophil function. Intestinal barrier function and nonsteroidal anti-inflammatory drug (NSAID)-induced injury were assessed in GM-CSF-null and NOD2-null mice. RESULTS: Median GM-CSF Ab levels increased from 0.4 microg/mL in control serum to 2.4 microg/mL in pediatric CD and 11.7 microg/mL in adult CD serum and were associated with ileal involvement (P<.001). Ileal location, duration of disease, and increased GM-CSF Ab levels were associated with stricturing/penetrating behavior (odds ratio, 2.2; P=.018). The positive and negative predictive values of GM-CSF Ab for stricturing/penetrating behavior were comparable with that of other IBD serum markers. CD patients with increased GM-CSF Ab had reduced neutrophil phagocytic capacity and increased accumulation of pSTAT3+ neutrophils in the affected ileum. GM-CSF-null mice and NOD2-null mice in which GM-CSF was neutralized had defects in mucosal barrier function and developed a transmural ileitis following NSAID exposure. CONCLUSIONS: GM-CSF regulates ileal homeostasis in CD and in mouse models. CD patients with increases in serum GM-CSF Ab might benefit from GM-CSF administration.


Subject(s)
Autoantibodies/blood , Crohn Disease/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Ileitis/immunology , Adult , Animals , Case-Control Studies , Child , Crohn Disease/blood , Crohn Disease/genetics , Disease Models, Animal , Disease Progression , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Humans , Ileitis/blood , Ileitis/genetics , Ileum/metabolism , Ileum/pathology , Kaplan-Meier Estimate , Male , Mice , Mice, Knockout , Neutrophils/metabolism , Neutrophils/pathology , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , STAT3 Transcription Factor/metabolism
16.
Inflamm Bowel Dis ; 14(4): 446-57, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18069684

ABSTRACT

BACKGROUND: While activation of the IL-6-dependent transcription factor signal transducer and activator of transcription 3 (STAT3) has been implicated in the pathogenesis of inflammatory bowel disease (IBD), a direct effect on mucosal gene expression and inflammation has not been shown. We hypothesized that a proinflammatory IL-6:STAT3-dependent biological network would be up regulated in pediatric-onset IBD patients, and would be associated with the severity of mucosal inflammation. METHODS: Patients with pediatric-onset IBD were enrolled at diagnosis and during therapy. Serum cytokine analysis was performed using Bioplex. STAT3 phosphorylation (pSTAT3) in peripheral blood leukocytes (PBLs) was assessed by flow cytometry. Immunohistochemistry of colonic mucosa was used to localize pSTAT3 and STAT3 target genes. Microarray analysis was used to determine RNA expression profiles from colon biopsies. RESULTS: Circulating IL-6 was upregulated in active IBD patients at diagnosis and during therapy. STAT3 activation was increased in PB granulocytes, IL-6-stimulated CD3(+)/CD4(+) lymphocytes, and affected colon biopsies of IBD patients. The frequency of pSTAT3+ PB granulocytes and colon epithelial and lamina propria cells was highly correlated with the degree of mucosal inflammation. Microarray and Ingenuity Systems bioinformatics analysis identified IL-6:STAT3-dependent biological networks upregulated in IBD patients which control leukocyte recruitment, HLA expression, angiogenesis, and tissue remodeling. CONCLUSIONS: A proinflammatory IL6:STAT3 biologic network is upregulated in active pediatric IBD patients at diagnosis and during therapy. Specific targeting of this network may be effective in reducing mucosal inflammation.


Subject(s)
Inflammatory Bowel Diseases/metabolism , Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Adolescent , Chemokine CXCL10/metabolism , Chemokine CXCL11/metabolism , Child , Colon/metabolism , Colon/pathology , Female , Gene Expression , Humans , Immunohistochemistry , Inflammation , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Interleukin-6/genetics , Intestinal Mucosa/metabolism , Leukocytes, Mononuclear/metabolism , Male , Oligonucleotide Array Sequence Analysis , Phosphorylation , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/genetics , Transcriptional Activation , Up-Regulation
17.
Mol Ther ; 12(5): 976-84, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16084127

ABSTRACT

Fanconi anemia (FA) is a rare autosomal recessive disorder that results from mutations in at least 11 different genes. Recent studies have demonstrated that clinical progression of the disease may be influenced by inter- and intragenic variations, emphasizing the importance of identifying the complementation groups. In the present study we have employed bicistronic retrovirus vectors that coexpress FA-specific cDNAs for complementation groups A, C, F, and G, together with the enhanced green fluorescence protein (EGFP), allowing for specific analysis of transduced EGFP+ cells within bulk cultures by flow cytometry. In addition, the assay relies on the correction of the characteristic FA-associated G2/M arrest after treatment of cells with DNA-damaging agents, which is analyzed by flow cytometry. Results obtained with this assay matched the complementation groups known for 12 control lymphoblast cell lines tested. We report here the results obtained for 48 FA patients with unknown complementation groups using this new assay. Complementation groups were identified for 24 patients. We have identified mutations in the genes corresponding to the assigned complementation group in 23 samples. This assay has now been established in a standardized fashion for complementation assignments in FA patients and the subsequent directing of rapid mutation analysis in those patients.


Subject(s)
DNA Mutational Analysis/methods , Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia/genetics , Retroviridae/genetics , Cell Line , Gene Transfer Techniques , Genetic Complementation Test , Genetic Vectors , Green Fluorescent Proteins , Humans , Mutation , Transfection
18.
Turk J Haematol ; 19(3): 391-7, 2002 Sep 05.
Article in English | MEDLINE | ID: mdl-27264975

ABSTRACT

The multi tumor suppressor genes MTS1 (CDKN2, p16INK4A) and MTS2 (CDKN1, p15INK4B) located at 9p21-22 are inactivated in some human cancers via several mechanisms including deletion and hypermethylation. In hematological malignancies, deletion of p16/p15 locus has been shown to be highly specific to lymphoid malignancies, and more particularly to T-cell acute lymphoblastic leukemia (TALL). We have investigated the deletion, methylation and p16 protein expression status of MTS1 in Tcell childhood acute lymphoblastic leukemia (19 cases) and cell lines[11]. On Southern blot homozygous deletions or hemizygous deletion with rearangement were detected in 4/19 T-ALL. The expression of p16 protein was not observed on Western blot in 4/15 T-ALL with intact p16 gene. The p16 gene was methylated 3/15 in T-ALL. Only one of three expressed p16 protein. The other 11/15 T-ALL had p16 protein expression but different level. Loss of MTS1 was observed in 3/11 cell lines. Cell line with MTS1 gene had p16 protein expression in 6/8. After treatment with the demethylating agent (5-AzoCyt) RD cell line showed p16 expression. This has not been observed with the other cell lines. Thus hypermethylation of MTS1 is rare in childhood T-ALL. Although inactivation of MTS1 by deletion is common in T-ALL and cell lines. Furthermore our data show that the p16 gene inactivation by hypermethylation and deletion may play a role in the leukemogenesis.

SELECTION OF CITATIONS
SEARCH DETAIL
...