Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
HGG Adv ; 5(2): 100279, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38389303

ABSTRACT

We designed a massively parallel reporter assay (MPRA) in an Epstein-Barr virus transformed B cell line to directly characterize the potential for histone post-translational modifications, i.e., histone quantitative trait loci (hQTLs), expression QTLs (eQTLs), and variants on systemic lupus erythematosus (SLE) and autoimmune (AI) disease risk haplotypes to modulate regulatory activity in an allele-dependent manner. Our study demonstrates that hQTLs, as a group, are more likely to modulate regulatory activity in an MPRA compared with other variant classes tested, including a set of eQTLs previously shown to interact with hQTLs and tested AI risk variants. In addition, we nominate 17 variants (including 11 previously unreported) as putative causal variants for SLE and another 14 for various other AI diseases, prioritizing these variants for future functional studies in primary and immortalized B cells. Thus, we uncover important insights into the mechanistic relationships among genotype, epigenetics, and gene expression in SLE and AI disease phenotypes.


Subject(s)
Epstein-Barr Virus Infections , Lupus Erythematosus, Systemic , Humans , Quantitative Trait Loci/genetics , Histones/genetics , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/genetics , Lupus Erythematosus, Systemic/genetics
2.
bioRxiv ; 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37645944

ABSTRACT

Objective: To systematically characterize the potential for histone post-translational modifications, i.e., histone quantitative trait loci (hQTLs), expression QTLs (eQTLs), and variants on systemic lupus erythematosus (SLE) and autoimmune (AI) disease risk haplotypes to modulate gene expression in an allele dependent manner. Methods: We designed a massively parallel reporter assay (MPRA) containing ~32K variants and transfected it into an Epstein-Barr virus transformed B cell line generated from an SLE case. Results: Our study expands our understanding of hQTLs, illustrating that epigenetic QTLs are more likely to contribute to functional mechanisms than eQTLs and other variant types, and a large proportion of hQTLs overlap transcription start sites (TSS) of noncoding RNAs. In addition, we nominate 17 variants (including 11 novel) as putative causal variants for SLE and another 14 for various other AI diseases, prioritizing these variants for future functional studies primary and immortalized B cells. Conclusion: We uncover important insights into the mechanistic relationships between genotype, epigenetics, gene expression, and SLE and AI disease phenotypes.

3.
Front Genet ; 13: 1011965, 2022.
Article in English | MEDLINE | ID: mdl-36199584

ABSTRACT

TNFAIP3/A20 is a prominent autoimmune disease risk locus that is correlated with hypomorphic TNFAIP3 expression and exhibits complex chromatin architecture with over 30 predicted enhancers. This study aimed to functionally characterize an enhancer ∼55 kb upstream of the TNFAIP3 promoter marked by the systemic lupus erythematosus (SLE) risk haplotype index SNP, rs10499197. Allele effects of rs10499197, rs58905141, and rs9494868 were tested by EMSA and/or luciferase reporter assays in immune cell types. Co-immunoprecipitation, ChIP-qPCR, and 3C-qPCR were performed on patient-derived EBV B cells homozygous for the non-risk or SLE risk TNFAIP3 haplotype to assess haplotype-specific effects on transcription factor binding and chromatin regulation at the TNFAIP3 locus. This study found that the TNFAIP3 locus has a complex chromatin regulatory network that spans ∼1M bp from the promoter region of IL20RA to the 3' untranslated region of TNFAIP3. Functional dissection of the enhancer demonstrated co-dependency of the RelA/p65 and CEBPB binding motifs that, together, increase IL20RA and IFNGR1 expression and decreased TNFAIP3 expression in the context of the TNFAIP3 SLE risk haplotype through dynamic long-range interactions up- and downstream. Examination of SNPs in linkage disequilibrium (D' = 1.0) with rs10499197 identified rs9494868 as a functional SNP with risk allele-specific increase in nuclear factor binding and enhancer activation in vitro. In summary, this study demonstrates that SNPs carried on the ∼109 kb SLE risk haplotype facilitate hypermorphic IL20RA and IFNGR1 expression, while suppressing TNFAIP3 expression, adding to the mechanistic potency of this critically important locus in autoimmune disease pathology.

4.
iScience ; 25(8): 104839, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35996584

ABSTRACT

Krüppel-like factor 4 (KLF4) is a transcription factor that has been proven necessary for both induction and maintenance of pluripotency and self-renewal. Whole-genome sequencing defined a unique mutation in KLF4 (KLF4K409Q) in human meningiomas. However, the molecular mechanism of this tumor-specific KLF4 mutation is unknown. Using genome-wide high-throughput and focused quantitative transcriptional approaches in human cell lines, primary meningeal cells, and meningioma tumor tissue, we found that a change in the evolutionarily conserved DNA-binding domain of KLF4 alters its DNA recognition preference, resulting in a shift in downstream transcriptional activity. In the KLF4K409Q-specific targets, the normally silent fibroblast growth factor 3 (FGF3) is activated. We demonstrated a neomorphic function of KLF4K409Q in stimulating FGF3 transcription through binding to its promoter and in using short tandem repeats (STRs) located within the locus as enhancers.

5.
Arthritis Rheumatol ; 74(1): 163-173, 2022 01.
Article in English | MEDLINE | ID: mdl-34279042

ABSTRACT

OBJECTIVE: Genetic variants spanning UBE2L3 are associated with increased expression of the UBE2L3-encoded E2 ubiquitin-conjugating enzyme H7 (UbcH7), which facilitates activation of proinflammatory NF-κB signaling and susceptibility to autoimmune diseases. We undertook this study to delineate how genetic variants carried on the UBE2L3/YDJC autoimmune risk haplotype function to drive hypermorphic UBE2L3 expression. METHODS: We used bioinformatic analyses, electrophoretic mobility shift assays, and luciferase reporter assays to identify and functionally characterize allele-specific effects of risk variants positioned in chromatin accessible regions of immune cells. Chromatin conformation capture with quantitative polymerase chain reaction (3C-qPCR), chromatin immunoprecipitation (ChIP)-qPCR, and small interfering RNA (siRNA) knockdown assays were performed on patient-derived Epstein-Barr virus-transformed B cells homozygous for the UBE2L3/YDJC nonrisk or risk haplotype to determine if the risk haplotype increases UBE2L3 expression by altering the regulatory chromatin architecture in the region. RESULTS: Of the 7 prioritized variants, 5 demonstrated allele-specific increases in nuclear protein binding affinity and regulatory activity. High-throughput sequencing of chromosome conformation capture coupled with ChIP (HiChIP) and 3C-qPCR uncovered a long-range interaction between the UBE2L3 promoter (rs140490, rs140491, rs11089620) and the downstream YDJC promoter (rs3747093) that was strengthened in the presence of the UBE2L3/YDJC risk haplotype, and correlated with the loss of CCCTC-binding factor (CTCF) and gain of YY1 binding at the risk alleles. Depleting YY1 by siRNA disrupted the long-range interaction between the 2 promoters and reduced UBE2L3 expression. CONCLUSION: The UBE2L3/YDJC autoimmune risk haplotype increases UBE2L3 expression through strengthening a YY1-mediated interaction between the UBE2L3 and YDJC promoters.


Subject(s)
Autoimmune Diseases/genetics , CCCTC-Binding Factor/physiology , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/physiology , YY1 Transcription Factor/physiology , Gene Expression Regulation , Genetic Variation , Haplotypes , Humans , Risk Factors
6.
Front Immunol ; 11: 567342, 2020.
Article in English | MEDLINE | ID: mdl-33363531

ABSTRACT

Sarcoidosis is a systemic inflammatory disease characterized by infiltration of immune cells into granulomas. Previous gene expression studies using heterogeneous cell mixtures lack insight into cell-type-specific immune dysregulation. We performed the first single-cell RNA-sequencing study of sarcoidosis in peripheral immune cells in 48 patients and controls. Following unbiased clustering, differentially expressed genes were identified for 18 cell types and bioinformatically assessed for function and pathway enrichment. Our results reveal persistent activation of circulating classical monocytes with subsequent upregulation of trafficking molecules. Specifically, classical monocytes upregulated distinct markers of activation including adhesion molecules, pattern recognition receptors, and chemokine receptors, as well as enrichment of immunoregulatory pathways HMGB1, mTOR, and ephrin receptor signaling. Predictive modeling implicated TGFß and mTOR signaling as drivers of persistent monocyte activation. Additionally, sarcoidosis T cell subsets displayed patterns of dysregulation. CD4 naïve T cells were enriched for markers of apoptosis and Th17/Treg differentiation, while effector T cells showed enrichment of anergy-related pathways. Differentially expressed genes in regulatory T cells suggested dysfunctional p53, cell death, and TNFR2 signaling. Using more sensitive technology and more precise units of measure, we identify cell-type specific, novel inflammatory and regulatory pathways. Based on our findings, we suggest a novel model involving four convergent arms of dysregulation: persistent hyperactivation of innate and adaptive immunity via classical monocytes and CD4 naïve T cells, regulatory T cell dysfunction, and effector T cell anergy. We further our understanding of the immunopathology of sarcoidosis and point to novel therapeutic targets.


Subject(s)
Gene Expression Profiling , Monocytes/immunology , Monocytes/metabolism , Sarcoidosis/etiology , Single-Cell Analysis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome , Apoptosis/genetics , Apoptosis/immunology , Biomarkers , Case-Control Studies , Cell Movement/genetics , Cell Movement/immunology , Clonal Anergy/genetics , Clonal Anergy/immunology , Disease Progression , Female , Humans , Immunophenotyping , Male , Models, Biological , Organ Specificity , Receptors, Antigen, T-Cell/metabolism , Sarcoidosis/diagnosis , Sarcoidosis/metabolism , Sarcoidosis/therapy , Signal Transduction
7.
Arthritis Rheumatol ; 72(5): 780-790, 2020 05.
Article in English | MEDLINE | ID: mdl-31804013

ABSTRACT

OBJECTIVE: Genetic variants in the region of tumor necrosis factor-induced protein 3-interacting protein 1 (TNIP1) are associated with autoimmune disease and reduced TNIP1 gene expression. The aim of this study was to define the functional genetic mechanisms driving TNIP1 hypomorphic expression imparted by the systemic lupus erythematosus-associated TNIP1 H1 risk haplotype. METHODS: Dual luciferase expression and electrophoretic mobility shift assays were used to evaluate the allelic effects of 11 risk variants on enhancer function and nuclear protein binding in immune cell line models (Epstein-Barr virus [EBV]-transformed human B cells, Jurkat cells, and THP-1 cells), left in a resting state or stimulated with phorbol 12-myristate 13-acetate/ionomycin. HiChIP was used to define the regulatory 3-dimensional (3-D) chromatin network of the TNIP1 haplotype by detecting in situ long-range DNA contacts associated with H3K27ac-marked chromatin in EBV B cells. Then, quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was used to determine the expression of genes within the 3-D chromatin network. RESULTS: Bioinformatics analyses of 50 single-nucleotide polymorphisms on the TNIP1 H1 risk haplotype identified 11 non-protein-coding variants with a high likelihood of influencing TNIP1 gene expression. Eight variants in EBV B cells, 5 in THP-1 cells, and 2 in Jurkat cells exhibited various allelic effects on enhancer activation, resulting in a cumulative suppressive effect on TNIP1 expression (net effect of risk variants -7.14 fold, -6.80 fold, and -2.44 fold, respectively; n > 3). Specifically, in EBV B cells, only 2 variants (rs10057690 and rs13180950) exhibited allele-specific loss of both enhancer activity and nuclear protein binding (each P < 0.01 relative to nonrisk alleles). In contrast, the rs10036748 risk allele reduced binding affinities of the transcriptional repressors basic helix-loop-helix family member 40/differentially expressed in chondrocytes 1 (bHLHe40/DEC1) (P < 0.05 relative to nonrisk alleles) and CREB-1 (P not significant) in EBV B cells, resulting in a gain of enhancer activity (P < 0.05). HiChIP and qRT-PCR analyses revealed that overall transcriptional repression of the TNIP1 haplotype extended to the neighboring genes DCTN4 and GMA2, both of which also showed decreased expression in the presence of the TNIP1 risk haplotype (P < 0.001 and P < 0.01, respectively, relative to the nonrisk haplotype); notably, it was found that these genes share a 3-D chromatin network. CONCLUSION: Hypomorphic TNIP1 expression results from the combined concordant and opposing effects of multiple risk variants carried on the TNIP1 risk haplotype, with the strongest regulatory effect in B lymphoid lineage cells. Furthermore, the TNIP1 risk haplotype effect extends to neighboring genes within a shared chromatin network.


Subject(s)
DNA-Binding Proteins/genetics , Lupus Erythematosus, Systemic/genetics , B-Lymphocytes , Chromatin , Gene Expression , Haplotypes , Humans , Risk Assessment
8.
Int J Cancer ; 147(4): 1086-1097, 2020 08 15.
Article in English | MEDLINE | ID: mdl-31845320

ABSTRACT

Current ovarian cancer maintenance therapy is limited by toxicity and no proven impact on overall survival. To study a maintenance strategy targeted at missense mutant p53, we hypothesized that the release of mutant p53 from mortalin inhibition by the SHetA2 drug combined with reactivation of mutant p53 with the PRIMA-1MET drug inhibits growth and tumor establishment synergistically in a mutant-p53 dependent manner. The Cancer Genome Atlas (TCGA) data and serous ovarian tumors were evaluated for TP53 and HSPA9/mortalin status. SHetA2 and PRIMA-1MET were tested in ovarian cancer cell lines and fallopian tube secretory epithelial cells using isobolograms, fluorescent cytometry, Western blots and ELISAs. Drugs were administered to mice after peritoneal injection of MESOV mutant p53 ovarian cancer cells and prior to tumor establishment, which was evaluated by logistic regression. Fifty-eight percent of TP53 mutations were missense and there were no mortalin mutations in TCGA high-grade serous ovarian cancers. Mortalin levels were sequentially increased in serous benign, borderline and carcinoma tumors. SHetA2 caused p53 nuclear and mitochondrial accumulation in cancer, but not in healthy, cells. Endogenous or exogenous mutant p53 increased SHetA2 resistance. PRIMA-1MET decreased this resistance and interacted synergistically with SHetA2 in mutant and wild type p53-expressing cell lines in association with elevated reactive oxygen species/ATP ratios. Tumor-free rates in animals were 0% (controls), 25% (PRIMA1MET ), 42% (SHetA2) and 67% (combination). SHetA2 (p = 0.004) and PRIMA1MET (p = 0.048) functioned additively in preventing tumor development with no observed toxicity. These results justify the development of SHetA2 and PRIMA-1MET alone and in combination for ovarian cancer maintenance therapy.


Subject(s)
Apoptosis/drug effects , Aza Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Chromans/pharmacology , HSP70 Heat-Shock Proteins/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Thiones/pharmacology , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Cell Line, Tumor , Female , HSP70 Heat-Shock Proteins/metabolism , Humans , Maintenance Chemotherapy/methods , Mice, Nude , Molecular Targeted Therapy/methods , Mutation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays/methods
9.
Nat Commun ; 9(1): 2905, 2018 07 25.
Article in English | MEDLINE | ID: mdl-30046115

ABSTRACT

Genetic variants can confer risk to complex genetic diseases by modulating gene expression through changes to the epigenome. To assess the degree to which genetic variants influence epigenome activity, we integrate epigenetic and genotypic data from lupus patient lymphoblastoid cell lines to identify variants that induce allelic imbalance in the magnitude of histone post-translational modifications, referred to herein as histone quantitative trait loci (hQTLs). We demonstrate that enhancer hQTLs are enriched on autoimmune disease risk haplotypes and disproportionately influence gene expression variability compared with non-hQTL variants in strong linkage disequilibrium. We show that the epigenome regulates HLA class II genes differently in individuals who carry HLA-DR3 or HLA-DR15 haplotypes, resulting in differential 3D chromatin conformation and gene expression. Finally, we identify significant expression QTL (eQTL) x hQTL interactions that reveal substructure within eQTL gene expression, suggesting potential implications for functional genomic studies that leverage eQTL data for subject selection and stratification.


Subject(s)
Chromatin/genetics , Haplotypes/genetics , Linkage Disequilibrium/genetics , Quantitative Trait Loci/genetics , Female , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Genotype , Humans , Male
10.
JCI Insight ; 1(8)2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27358913

ABSTRACT

CD4+ T cells predominate in salivary gland (SG) inflammatory lesions in Sjögren's syndrome (SS). However, their antigen specificity, degree of clonal expansion, and relationship to clinical disease features remain unknown. We used multiplex reverse-transcriptase PCR to amplify paired T cell receptor α (TCRα) and ß transcripts of single CD4+CD45RA- T cells from SG and peripheral blood (PB) of 10 individuals with primary SS, 9 of whom shared the HLA DR3/DQ2 risk haplotype. TCRα and ß sequences were obtained from a median of 91 SG and 107 PB cells per subject. The degree of clonal expansion and frequency of cells expressing two productively rearranged α genes were increased in SG versus PB. Expanded clones from SG exhibited complementary-determining region 3 (CDR3) sequence similarity both within and among subjects, suggesting antigenic selection and shared antigen recognition. CDR3 similarities were shared among expanded clones from individuals discordant for canonical Ro and La autoantibodies, suggesting recognition of alternative SG antigen(s). The extent of SG clonal expansion correlated with reduced saliva production and increased SG fibrosis, linking expanded SG T cells with glandular dysfunction. Knowledge of paired TCRα and ß sequences enables further work toward identification of target antigens and development of novel therapies.

11.
Mol Cell Biol ; 35(10): 1763-76, 2015 May.
Article in English | MEDLINE | ID: mdl-25755281

ABSTRACT

Molecules involved in WNT/ß-catenin signaling show specific spatiotemporal expression and play vital roles in myogenesis; however, it is still largely unknown how WNT/ß-catenin signaling regulates each step of myogenesis. Here, we show that WNT/ß-catenin signaling can control diverse biological processes of myogenesis by regulating step-specific molecules. In order to identify the temporally specific roles of WNT/ß-catenin signaling molecules in muscle development and homeostasis, we used in vitro culture systems for both primary mouse myoblasts and C2C12 cells, which can differentiate into myofibers. We found that a blockade of WNT/ß-catenin signaling in the proliferating cells decreases proliferation activity, but does not induce cell death, through the regulation of genes cyclin A2 (Ccna2) and cell division cycle 25C (Cdc25c). During muscle differentiation, the inhibition of WNT/ß-catenin signaling blocks myoblast fusion through the inhibition of the Fermitin family homolog 2 (Fermt2) gene. Blocking WNT/ß-catenin signaling in the well-differentiated myofibers results in the failure of maintenance of their structure by disruption of cadherin/ß-catenin/actin complex formation, which plays a crucial role in connecting a myofiber's cytoskeleton to the surrounding extracellular matrix. Thus, our results indicate that WNT/ß-catenin signaling can regulate multiple steps of myogenesis, including cell proliferation, myoblast fusion, and homeostasis, by targeting step-specific molecules.


Subject(s)
Muscle Development , Myoblasts/cytology , Wnt Signaling Pathway , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Cells, Cultured , Cyclin A2/genetics , Cyclin A2/metabolism , Homeostasis , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Myoblasts/metabolism , Tongue/cytology , Wnt Signaling Pathway/drug effects , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
12.
Hum Mol Genet ; 23(1): 182-93, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-23975680

ABSTRACT

Mutations in transforming growth factor beta (TGFß) receptor type II (TGFBR2) cause Loeys-Dietz syndrome, characterized by craniofacial and cardiovascular abnormalities. Mice with a deletion of Tgfbr2 in cranial neural crest cells (Tgfbr2(fl/fl);Wnt1-Cre mice) develop cleft palate as the result of abnormal TGFß signaling activation. However, little is known about metabolic processes downstream of TGFß signaling during palatogenesis. Here, we show that Tgfbr2 mutant palatal mesenchymal cells spontaneously accumulate lipid droplets, resulting from reduced lipolysis activity. Tgfbr2 mutant palatal mesenchymal cells failed to respond to the cell proliferation stimulator sonic hedgehog, derived from the palatal epithelium. Treatment with p38 mitogen-activated protein kinase (MAPK) inhibitor or telmisartan, a modulator of p38 MAPK activation and lipid metabolism, blocked abnormal TGFß-mediated p38 MAPK activation, restoring lipid metabolism and cell proliferation activity both in vitro and in vivo. Our results highlight the influence of alternative TGFß signaling on lipid metabolic activities, as well as how lipid metabolic defects can affect cell proliferation and adversely impact palatogenesis. This discovery has broader implications for the understanding of metabolic defects and potential prevention of congenital birth defects.


Subject(s)
Benzimidazoles/pharmacology , Benzoates/pharmacology , Cleft Palate/drug therapy , Lipid Metabolism/drug effects , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Cell Line , Cell Proliferation/drug effects , Cleft Palate/embryology , Cleft Palate/pathology , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Telmisartan , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Biol Chem ; 288(41): 29760-70, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23950180

ABSTRACT

Microglossia is a congenital birth defect in humans and adversely impacts quality of life. In vertebrates, tongue muscle derives from the cranial mesoderm, whereas tendons and connective tissues in the craniofacial region originate from cranial neural crest (CNC) cells. Loss of transforming growth factor ß (TGFß) type II receptor in CNC cells in mice (Tgfbr2(fl/fl);Wnt1-Cre) causes microglossia due to a failure of cell-cell communication between cranial mesoderm and CNC cells during tongue development. However, it is still unclear how TGFß signaling in CNC cells regulates the fate of mesoderm-derived myoblasts during tongue development. Here we show that activation of the cytoplasmic and nuclear tyrosine kinase 1 (ABL1) cascade in Tgfbr2(fl/fl);Wnt1-Cre mice results in a failure of CNC-derived cell differentiation followed by a disruption of TGFß-mediated induction of growth factors and reduction of myogenic cell proliferation and differentiation activities. Among the affected growth factors, the addition of fibroblast growth factor 4 (FGF4) and neutralizing antibody for follistatin (FST; an antagonist of bone morphogenetic protein (BMP)) could most efficiently restore cell proliferation, differentiation, and organization of muscle cells in the tongue of Tgfbr2(fl/fl);Wnt1-Cre mice. Thus, our data indicate that CNC-derived fibroblasts regulate the fate of mesoderm-derived myoblasts through TGFß-mediated regulation of FGF and BMP signaling during tongue development.


Subject(s)
Muscles/metabolism , Neural Crest/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Tongue/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Female , Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 4/metabolism , Fibroblast Growth Factor 4/pharmacology , Follistatin/genetics , Follistatin/metabolism , Follistatin/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Immunoblotting , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Muscles/embryology , Neural Crest/cytology , Neural Crest/embryology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Skull/embryology , Skull/metabolism , Tongue/cytology , Tongue/embryology , Transforming Growth Factor beta/pharmacology
14.
Development ; 140(6): 1220-30, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23406900

ABSTRACT

Cleft palate is one of the most common human birth defects and is associated with multiple genetic and environmental risk factors. Although mutations in the genes encoding transforming growth factor beta (TGFß) signaling molecules and interferon regulatory factor 6 (Irf6) have been identified as genetic risk factors for cleft palate, little is known about the relationship between TGFß signaling and IRF6 activity during palate formation. Here, we show that TGFß signaling regulates expression of Irf6 and the fate of the medial edge epithelium (MEE) during palatal fusion in mice. Haploinsufficiency of Irf6 in mice with basal epithelial-specific deletion of the TGFß signaling mediator Smad4 (Smad4(fl/fl);K14-Cre;Irf6(+/R84C)) results in compromised p21 expression and MEE persistence, similar to observations in Tgfbr2(fl/fl);K14-Cre mice, although the secondary palate of Irf6(+/R84C) and Smad4(fl/fl);K14-Cre mice form normally. Furthermore, Smad4(fl/fl);K14-Cre;Irf6(+/R84C) mice show extra digits that are consistent with abnormal toe and nail phenotypes in individuals with Van der Woude and popliteal pterygium syndromes, suggesting that the TGFß/SMAD4/IRF6 signaling cascade might be a well-conserved mechanism in regulating multiple organogenesis. Strikingly, overexpression of Irf6 rescued p21 expression and MEE degeneration in Tgfbr2(fl/fl);K14-Cre mice. Thus, IRF6 and SMAD4 synergistically regulate the fate of the MEE, and TGFß-mediated Irf6 activity is responsible for MEE degeneration during palatal fusion in mice.


Subject(s)
Epistasis, Genetic , Interferon Regulatory Factors/genetics , Palate/embryology , Smad4 Protein/genetics , Transforming Growth Factor beta/pharmacology , Animals , Animals, Newborn , Cell Fusion , Cells, Cultured , Embryo, Mammalian , Epistasis, Genetic/drug effects , Epistasis, Genetic/physiology , Female , Humans , Interferon Regulatory Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organogenesis/drug effects , Organogenesis/genetics , Palate/drug effects , Palate/metabolism , Pregnancy , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism
15.
J Cell Biochem ; 114(4): 796-807, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23060211

ABSTRACT

Nonsyndromic orofacial clefts are common birth defects whose etiology is influenced by complex genetic and environmental factors and gene-environment interactions. Although these risk factors are not yet fully elucidated, it is known that alterations in transforming growth factor-beta (TGFß) signaling can cause craniofacial abnormalities, including cleft palate, in mammals. To elucidate the downstream targets of TGFß signaling in palatogenesis, we analyzed the gene expression profiles of Tgfbr2(fl/fl) ;Wnt1-Cre mouse embryos with cleft palate and other craniofacial deformities resulting from the targeted inactivation of the Tgfbr2 gene in their cranial neural crest (CNC) cells. Relative to controls, palatal tissues obtained from Tgfbr2(fl/fl) ;Wnt1-Cre mouse embryos at embryonic day 14.5 (E14.5) of gestation have a robust gene expression signature reflective of known defects in CNC-derived mesenchymal cell proliferation. Groups of differentially expressed genes (DEGs) were involved in diverse cellular processes and components associated with orofacial clefting, including the extracellular matrix, cholesterol metabolism, ciliogenesis, and multiple signaling pathways. A subset of the DEGs are known or suspected to be associated with an increased risk of orofacial clefting in humans and/or genetically engineered mice. Based on bioinformatics analyses, we highlight the functional relationships among differentially expressed transcriptional regulators of palatogenesis as well as transcriptional factors not previously associated with this process. We suggest that gene expression profiling studies of mice with TGFß signaling defects provide a valuable approach for identifying candidate mechanisms by which this pathway controls cell fate during palatogenesis and its role in the etiology of human craniofacial abnormalities.


Subject(s)
Cleft Palate/pathology , Gene Expression Profiling/methods , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Binding Sites , Cell Differentiation , Cell Proliferation , Cleft Palate/embryology , Cleft Palate/metabolism , Computational Biology/methods , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Gene Silencing , Male , Mice , Mice, Inbred C57BL , Nucleotide Motifs , Palate/embryology , Palate/metabolism , Palate/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/genetics , Wnt1 Protein/genetics , Wnt1 Protein/metabolism
16.
AMIA Annu Symp Proc ; 2010: 632-6, 2010 Nov 13.
Article in English | MEDLINE | ID: mdl-21347055

ABSTRACT

Mass spectrometry proteomic profiling has potential to be a useful clinical screening tool. One obstacle is providing a standardized method for preprocessing the noisy raw data. We have developed a system for automatically determining a set of preprocessing methods among several candidates. Our system's automated nature relieves the analyst of the need to be knowledgeable about which methods to use on any given dataset. Each stage of preprocessing is approached with many competing methods. We introduce metrics which are used to balance each method's attempts to correct noise versus preserving valuable discriminative information. We demonstrate the benefit of our preprocessing system on several SELDI and MALDI mass spectrometry datasets. Downstream classification is improved when using our system to preprocess the data.


Subject(s)
Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Algorithms , Gene Expression Profiling , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...