Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Nov 14.
Article in English | MEDLINE | ID: mdl-38014069

ABSTRACT

A nucleotide repeat expansion (NRE) in the first annotated intron of the C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). While C9 NRE-containing RNAs can be translated into several toxic dipeptide repeat proteins, how an intronic NRE can assess the translation machinery in the cytoplasm remains unclear. By capturing and sequencing NRE-containing RNAs from patient-derived cells, we found that C9 NRE was exonized by the usage of downstream 5' splice sites and exported from the nucleus in a variety of spliced mRNA isoforms. C9ORF72 aberrant splicing was substantially elevated in both C9 NRE+ motor neurons and human brain tissues. Furthermore, NREs above the pathological threshold were sufficient to activate cryptic splice sites in reporter mRNAs. In summary, our results revealed a crucial and potentially widespread role of repeat-induced aberrant splicing in the biogenesis, localization, and translation of NRE-containing RNAs.

2.
J Am Chem Soc ; 145(37): 20273-20288, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37695732

ABSTRACT

The cGMP-AMP Synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway plays a critical role in sensing dsDNA localized to the cytosol, resulting in the activation of a robust inflammatory response. While cGAS-STING signaling is essential for antiviral immunity, aberrant STING activation is observed in amyotrophic lateral sclerosis (ALS), lupus, and autoinflammatory diseases such as Aicardi-Goutières syndrome (AGS) and STING associated vasculopathy with onset in infancy (SAVI). Significant efforts have therefore focused on the development of STING inhibitors. In a concurrent submission, we reported that BB-Cl-amidine inhibits STING-dependent signaling in the nanomolar range, both in vitro and in vivo. Considering this discovery, we sought to generate analogs with higher potency and proteome-wide selectivity. Herein, we report the development of LB244, which displays nanomolar potency and inhibits STING signaling with markedly enhanced proteome-wide selectivity. Moreover, LB244 mirrored the efficacy of BB-Cl-amidine in vivo. In summary, our data identify novel chemical entities that inhibit STING signaling and provide a scaffold for the development of therapeutics for treating STING-dependent inflammatory diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Autoimmune Diseases of the Nervous System , Humans , Proteome , Antiviral Agents , Cyclic GMP , Nucleotidyltransferases
3.
Hepatol Commun ; 7(10)2023 10 01.
Article in English | MEDLINE | ID: mdl-37756045

ABSTRACT

BACKGROUND: Genome-wide association studies (GWAS) have identified 30 risk loci for primary sclerosing cholangitis (PSC). Variants within these loci are found predominantly in noncoding regions of DNA making their mechanisms of conferring risk hard to define. Epigenomic studies have shown noncoding variants broadly impact regulatory element activity. The possible association of noncoding PSC variants with regulatory element activity has not been studied. We aimed to (1) determine if the noncoding risk variants in PSC impact regulatory element function and (2) if so, assess the role these regulatory elements have in explaining the genetic risk for PSC. METHODS: Available epigenomic datasets were integrated to build a comprehensive atlas of cell type-specific regulatory elements, emphasizing PSC-relevant cell types. RNA-seq and ATAC-seq were performed on peripheral CD4+ T cells from 10 PSC patients and 11 healthy controls. Computational techniques were used to (1) study the enrichment of PSC-risk variants within regulatory elements, (2) correlate risk genotype with differences in regulatory element activity, and (3) identify regulatory elements differentially active and genes differentially expressed between PSC patients and controls. RESULTS: Noncoding PSC-risk variants are strongly enriched within immune-specific enhancers, particularly ones involved in T-cell response to antigenic stimulation. In total, 250 genes and >10,000 regulatory elements were identified that are differentially active between patients and controls. CONCLUSIONS: Mechanistic effects are proposed for variants at 6 PSC-risk loci where genotype was linked with differential T-cell regulatory element activity. Regulatory elements are shown to play a key role in PSC pathophysiology.


Subject(s)
Cholangitis, Sclerosing , Genome-Wide Association Study , Humans , Cholangitis, Sclerosing/genetics , Chromatin Immunoprecipitation Sequencing , Genotype
4.
Proc Natl Acad Sci U S A ; 120(33): e2305420120, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37549268

ABSTRACT

Stimulator of interferon genes (STING) is an essential adaptor protein required for the inflammatory response to cytosolic DNA. dsDNA activates cGAS to generate cGAMP, which binds and activates STING triggering a conformational change, oligomerization, and the IRF3- and NFκB-dependent transcription of type I Interferons (IFNs) and inflammatory cytokines, as well as the activation of autophagy. Aberrant activation of STING is now linked to a growing number of both rare as well as common chronic inflammatory diseases. Here, we identify and characterize a potent small-molecule inhibitor of STING. This compound, BB-Cl-amidine inhibits STING signaling and production of type I IFNs, IFN-stimulated genes (ISGs) and NFκB-dependent cytokines, but not other pattern recognition receptors. In vivo, BB-Cl-amidine alleviated pathology resulting from accrual of cytosolic DNA in Trex-1 mutant mice. Mechanistically BB-Cl-amidine inhibited STING oligomerization through modification of Cys148. Collectively, our work uncovers an approach to inhibit STING activation and highlights the potential of this strategy for the treatment of STING-driven inflammatory diseases.


Subject(s)
Interferon Type I , Membrane Proteins , Mice , Animals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Signal Transduction/physiology , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Interferon Type I/metabolism , NF-kappa B/metabolism , DNA
5.
Proc Natl Acad Sci U S A ; 120(5): e2213777120, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36693106

ABSTRACT

The accrual of cytosolic DNA leads to transcription of type I IFNs, proteolytic maturation of the IL-1 family of cytokines, and pyroptotic cell death. Caspase-1 cleaves pro-IL1ß to generate mature bioactive cytokine and gasdermin D which facilitates IL-1 release and pyroptotic cell death. Absent in melanoma-2 (AIM2) is a sensor of dsDNA leading to caspase-1 activation, although in human monocytes, cGAS-STING acting upstream of NLRP3 mediates the dsDNA-activated inflammasome response. In healthy human keratinocytes, AIM2 is not expressed yet caspase-1 is activated by the synthetic dsDNA mimetic poly(dA:dT). Here, we show that this response is not mediated by either AIM2 or the cGAS-STING-NLRP3 pathway and is instead dependent on NLRP1. Poly(dA:dT) is unique in its ability to activate NLRP1, as conventional linear dsDNAs fail to elicit NLRP1 activation. DsRNA was recently shown to activate NLRP1 and prior work has shown that poly(dA:dT) is transcribed into an RNA intermediate that stimulates the RNA sensor RIG-I. However, poly(dA:dT)-dependent RNA intermediates are insufficient to activate NLRP1. Instead, poly(dA:dT) results in oxidative nucleic acid damage and cellular stress, events which activate MAP3 kinases including ZAKα that converge on p38 to activate NLRP1. Collectively, this work defines a new activator of NLRP1, broadening our understanding of sensors that recognize poly(dA:dT) and advances the understanding of the immunostimulatory potential of this potent adjuvant.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Cytokines/metabolism , DNA/metabolism , Caspase 1/metabolism , RNA/metabolism , Keratinocytes/metabolism , Interleukin-1/metabolism , NLR Proteins/metabolism
6.
Immunity ; 55(8): 1340-1342, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35947978

ABSTRACT

During infection, pore-forming proteins rapidly initiate cell lysis, but specialized processes like epithelial extrusion need additional time to occur in parallel. In a recent issue of Nature, Nozaki et al. (2022) report that caspase-7 promotes acid shingomyelinase (ASM)-mediated membrane repair of gasdermin and perforin pores to delay cell death.


Subject(s)
Caspase 7 , Cell Membrane/metabolism , Perforin/metabolism , Pore Forming Cytotoxic Proteins
9.
J Rehabil Med Clin Commun ; 4: 1000056, 2021.
Article in English | MEDLINE | ID: mdl-33884156

ABSTRACT

OBJECTIVE: Low back pain caused by osteoporosisinduced thoracolumbar vertebral compression fractures is a common debilitating disorder. The aims of this study were to determine the accuracy and efficacy of spinal dorsal ramus injection and radiofrequency neurolysis for pain reduction in patients with this condition. METHODS: This study was a retrospective chart review of 46 patients with low back pain caused by osteoporosis-induced thoracolumbar vertebral compression fractures. All patients had been treated with spinal dorsal ramus injection with mixed Sensorcaine (Fresenius Kabi, USA) and Depo-Medrol (Pfizer, USA). In some patients further treatment with radiofrequency neurolysis had been required after the initial injection wore off. RESULTS: Out of a total of 46 patients, 45 (97.7%) had ≥ 50% reduction in low back pain immediately after injection. After the initial injection wore off, 18 patients remained pain free and 27 required radiofrequency neurolysis. The follow-up period ranged from 60 to 1,440 days (mean 335 days). The intensity of low back pain decreased from 7.09 ± 0.84 (numerical pain scale of 0-10) before treatment to 1.39 ± 1.51 immediately after the injection, and to 0.96 ± 1.36 at the last office visit. CONCLUSION: Spinal dorsal ramus injection and radiofrequency neurolysis are effective and accurate therapies for low back pain caused by osteoporosis-induced thoracolumbar vertebral compression fractures.

10.
Elife ; 102021 01 27.
Article in English | MEDLINE | ID: mdl-33501914

ABSTRACT

Lymphoblastoid cell lines (LCLs) are generated by transforming primary B cells with Epstein-Barr virus (EBV) and are used extensively as model systems in viral oncology, immunology, and human genetics research. In this study, we characterized single-cell transcriptomic profiles of five LCLs and present a simple discrete-time simulation to explore the influence of stochasticity on LCL clonal evolution. Single-cell RNA sequencing (scRNA-seq) revealed substantial phenotypic heterogeneity within and across LCLs with respect to immunoglobulin isotype; virus-modulated host pathways involved in survival, activation, and differentiation; viral replication state; and oxidative stress. This heterogeneity is likely attributable to intrinsic variance in primary B cells and host-pathogen dynamics. Stochastic simulations demonstrate that initial primary cell heterogeneity, random sampling, time in culture, and even mild differences in phenotype-specific fitness can contribute substantially to dynamic diversity in populations of nominally clonal cells.


Subject(s)
Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Transcriptome , B-Lymphocytes/physiology , Cell Line , Humans , RNA-Seq , Single-Cell Analysis
12.
Biochemistry ; 59(50): 4755-4765, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33272017

ABSTRACT

In the MAPK pathway, an oncogenic V600E mutation in B-Raf kinase causes the enzyme to be constitutively active, leading to aberrantly high phosphorylation levels of its downstream effectors, MEK and ERK kinases. The V600E mutation in B-Raf accounts for more than half of all melanomas and ∼3% of all cancers, and many drugs target the ATP binding site of the enzyme for its inhibition. Because B-Raf can develop resistance against these drugs and such drugs can induce paradoxical activation, drugs that target allosteric sites are needed. To identify other potential drug targets, we generated and kinetically characterized an active form of B-RafV600E expressed using a bacterial expression system. In doing so, we identified an α-helix on B-Raf, found at the B-Raf-MEK interface, that is critical for their interaction and the oncogenic activity of B-RafV600E. We assessed the binding between B-Raf mutants and MEK using pull downs and biolayer interferometry and assessed phosphorylation levels of MEK in vitro and in cells as well as its downstream target ERK to show that mutating certain residues on this α-helix is detrimental to binding and downstream activity. Our results suggest that this B-Raf α-helix binding site on MEK could be a site to target for drug development to treat B-RafV600E-induced melanomas.


Subject(s)
MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 1/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , Allosteric Site , Amino Acid Sequence , Drug Discovery , Drug Resistance, Neoplasm , HEK293 Cells , Humans , In Vitro Techniques , Kinetics , MAP Kinase Kinase 1/genetics , MAP Kinase Signaling System , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Phosphorylation , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins B-raf/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity
13.
Retrovirology ; 17(1): 28, 2020 08 31.
Article in English | MEDLINE | ID: mdl-32867805

ABSTRACT

BACKGROUND: Antiretroviral therapy (ART) can mitigate the morbidity and mortality caused by the human immunodeficiency virus (HIV). Successful development of ART can be accelerated by accurate structural and biochemical data on targets and their responses to inhibitors. One important ART target, HIV integrase (IN), has historically been studied in vitro in a modified form adapted to bacterial overexpression, with a methionine or a longer fusion protein sequence at the N-terminus. In contrast, IN present in viral particles is produced by proteolytic cleavage of the Pol polyprotein, which leaves a phenylalanine at the N-terminus (IN 1F). Inspection of available structures suggested that added residues on the N-terminus might disrupt proper protein folding and formation of multimeric complexes. RESULTS: We purified HIV-1 IN 1F1-212 and solved its structure at 2.4 Å resolution, which showed extension of an N-terminal helix compared to the published structure of IN1-212. Full-length IN 1F showed increased in vitro catalytic activity in assays of coupled joining of the two viral DNA ends compared to two IN variants containing additional N-terminal residues. IN 1F was also altered in its sensitivity to inhibitors, showing decreased sensitivity to the strand-transfer inhibitor raltegravir and increased sensitivity to allosteric integrase inhibitors. In solution, IN 1F exists as monomers and dimers, in contrast to other IN preparations which exist as higher-order oligomers. CONCLUSIONS: The structural, biochemical, and biophysical characterization of IN 1F reveals the conformation of the native HIV-1 IN N-terminus and accompanying unique biochemical and biophysical properties. IN 1F thus represents an improved reagent for use in integration reactions in vitro and the development of antiretroviral agents.


Subject(s)
HIV Integrase/chemistry , HIV Integrase/metabolism , HIV-1/enzymology , Allosteric Regulation/drug effects , Crystallography, X-Ray , DNA, Viral/metabolism , HIV Integrase/genetics , HIV Integrase Inhibitors/pharmacology , HIV-1/chemistry , Humans , Phenylalanine , Protein Conformation , Protein Folding , Raltegravir Potassium/pharmacology , Structure-Activity Relationship
14.
Front Immunol ; 11: 1529, 2020.
Article in English | MEDLINE | ID: mdl-32765530

ABSTRACT

Understanding affinity maturation of antibodies that can target many variants of HIV-1 is important for vaccine development. While the antigen-binding site of antibodies is known to mutate throughout the co-evolution of antibodies and viruses in infected individuals, the roles of the mutations in the antibody framework region are not well understood. Throughout affinity maturation, the CH103 broadly neutralizing antibody lineage, from an individual designated CH505, altered the orientation of one of its antibody variable domains. The change in orientation was a response to insertions in the variable loop 5 (V5) of the HIV envelope. In this study, we generated CH103 lineage antibody variants in which residues in the variable domain interface were mutated, and measured the binding to both autologous and heterologous HIV-1 envelopes. Our data show that very few mutations in an early intermediate antibody of the lineage can improve binding toward both autologous and heterologous HIV-1 envelopes. We also crystallized an antibody mutant to show that framework mutations alone can result in a shift in relative orientations of the variable domains. Taken together, our results demonstrate the functional importance of residues located outside the antigen-binding site in affinity maturation.


Subject(s)
Antibody Affinity/genetics , HIV Antibodies/genetics , HIV Antibodies/immunology , HIV Infections/genetics , HIV Infections/immunology , HIV-1/immunology , Immunoglobulin Variable Region/genetics , Mutation , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Epitopes/chemistry , Epitopes/immunology , HIV Antibodies/chemistry , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/immunology , HIV Infections/virology , Humans , Immunoglobulin Variable Region/chemistry , Models, Molecular , Protein Binding , Protein Conformation , Structure-Activity Relationship
15.
Nat Commun ; 11(1): 3354, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620797

ABSTRACT

Expansion of an intronic (GGGGCC)n repeat region within the C9orf72 gene is a main cause of familial amyotrophic lateral sclerosis and frontotemporal dementia (c9ALS/FTD). A hallmark of c9ALS/FTD is the accumulation of misprocessed RNAs, which are often targets of cellular RNA surveillance. Here, we show that RNA decay mechanisms involving upstream frameshift 1 (UPF1), including nonsense-mediated decay (NMD), are inhibited in c9ALS/FTD brains and in cultured cells expressing either of two arginine-rich dipeptide repeats (R-DPRs), poly(GR) and poly(PR). Mechanistically, although R-DPRs cause the recruitment of UPF1 to stress granules, stress granule formation is independent of NMD inhibition. Instead, NMD inhibition is primarily a result from global translational repression caused by R-DPRs. Overexpression of UPF1, but none of its NMD-deficient mutants, enhanced the survival of neurons treated by R-DPRs, suggesting that R-DPRs cause neurotoxicity in part by inhibiting cellular RNA surveillance.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , Frontotemporal Dementia/genetics , Nonsense Mediated mRNA Decay , RNA Helicases/metabolism , Trans-Activators/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Line, Tumor , Cell Survival/genetics , DNA Repeat Expansion , Datasets as Topic , Embryo, Mammalian , Female , Frontal Lobe/pathology , Frontotemporal Dementia/pathology , Humans , Introns/genetics , Mice , Neurons/metabolism , Neurons/pathology , Primary Cell Culture , Protein Biosynthesis , RNA, Messenger/metabolism , RNA-Seq , Trans-Activators/genetics
16.
Methods Mol Biol ; 1856: 283-296, 2018.
Article in English | MEDLINE | ID: mdl-30178259

ABSTRACT

Pyrosequencing, a real-time sequencing technology, is considered a "gold standard" for quantitative allele quantification at single base resolution. Quantitative bisulfite Pyrosequencing determines DNA methylation level by analyzing artificial "C/T" SNPs at CpG sites within a specific Pyrosequencing assay. The bisulfite Pyrosequencing methylation assay design is DNA strand specific and the primer design should not contain any CpG sites and should be free of high-frequency mutations. Additionally Pyrosequencing assays must be tested for preferential amplification during bisulfite PCR to ensure the sequencing quantification accuracy and reproducibility. Pyrosequencing analysis gives a reproducible measurement of average methylation at several CpG sites within the Pyrosequencing assay directly from a PCR product, rapidly and accurately for many samples at a time. It is therefore well suited for clinical research, validation of whole-genome methylation screening results, and global methylation analysis using repetitive elements including LINE-1, Alu, and Sat2. Pyrosequencing reproducibility and accuracy result in low measurement variance, thereby increasing the likelihood of early detection of small changes in methylation levels that may become apparent in response to treatment. For example, the high reproducibility of the LINE-1 assay is important for detecting the relatively small daily changes in methylation levels associated with hypomethylation. This enables detection of differences in patterns between normal and disease tissue such as in tumor suppresser genes, and to determine global methylation changes in response drug treatments. Relatively low cost and easy automation allows the researcher to increase the experiment's sample population to detect trends that would otherwise not have a sufficient sampling basis for statistical significance.


Subject(s)
DNA Methylation , Epigenomics , Sequence Analysis, DNA , CpG Islands , Epigenomics/methods , Genome-Wide Association Study/methods , Humans , Polymerase Chain Reaction , Reproducibility of Results , Sequence Analysis, DNA/methods
17.
AIDS Res Hum Retroviruses ; 34(9): 760-768, 2018 09.
Article in English | MEDLINE | ID: mdl-29984587

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) is a rapidly evolving pathogen that causes acquired immunodeficiency syndrome (AIDS) in humans. There are ∼30-35 million people infected with HIV around the world, and ∼25 million have died since the first reported cases in 1981. In addition, each year 2-3 million people become newly infected, and >1 million die of AIDS. An HIV-1 vaccine would help halt an AIDS pandemic, and efforts to develop a vaccine have focused on targeting the HIV-1 envelope, Env, found on the surface of the virus. A number of chronically infected individuals have been shown to produce antibodies, called broadly neutralizing antibodies (bnAbs), that target many strains of HIV-1 by binding to Env, thus suggesting promise for HIV-1 vaccine development. BnAbs take years to develop, and have a number of traits that inhibit their production; thus, a number of researchers are trying to understand the pathways that result in bnAb production, so that they can be elicited more rapidly by vaccination. This review discusses results and implications from two HIV-1-infected individuals studied longitudinally who produced bnAbs against two different sites on HIV-1 Env, and immunization studies that used Envs derived from those individuals.


Subject(s)
Acquired Immunodeficiency Syndrome/immunology , HIV Antibodies/immunology , HIV Infections/immunology , HIV-1/immunology , AIDS Vaccines/immunology , Acquired Immunodeficiency Syndrome/prevention & control , Animals , Antibodies, Neutralizing/immunology , HIV Infections/prevention & control , Humans , Immunization/methods , Vaccination/methods , env Gene Products, Human Immunodeficiency Virus/immunology
18.
Nat Commun ; 9(1): 1111, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29549260

ABSTRACT

HIV-1 envelope (Env) mimetics are candidate components of prophylactic vaccines and potential therapeutics. Here we use a synthetic V3-glycopeptide ("Man9-V3") for structural studies of an HIV Env third variable loop (V3)-glycan directed, broadly neutralizing antibody (bnAb) lineage ("DH270"), to visualize the epitope on Env and to study how affinity maturation of the lineage proceeded. Unlike many previous V3 mimetics, Man9-V3 encompasses two key features of the V3 region recognized by V3-glycan bnAbs-the conserved GDIR motif and the N332 glycan. In our structure of an antibody fragment of a lineage member, DH270.6, in complex with the V3 glycopeptide, the conformation of the antibody-bound glycopeptide conforms closely to that of the corresponding segment in an intact HIV-1 Env trimer. An additional structure identifies roles for two critical mutations in the development of breadth. The results suggest a strategy for use of a V3 glycopeptide as a vaccine immunogen.


Subject(s)
Antibodies, Neutralizing/immunology , Gene Products, env/chemistry , Gene Products, env/immunology , HIV Antibodies/immunology , HIV Infections/virology , HIV-1/immunology , Amino Acid Motifs , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Gene Products, env/genetics , HIV Infections/immunology , HIV-1/chemistry , HIV-1/genetics , Humans , Models, Molecular , Mutation
19.
Genes Dev ; 29(12): 1298-315, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26080815

ABSTRACT

Mutant p53 impacts the expression of numerous genes at the level of transcription to mediate oncogenesis. We identified vascular endothelial growth factor receptor 2 (VEGFR2), the primary functional VEGF receptor that mediates endothelial cell vascularization, as a mutant p53 transcriptional target in multiple breast cancer cell lines. Up-regulation of VEGFR2 mediates the role of mutant p53 in increasing cellular growth in two-dimensional (2D) and three-dimensional (3D) culture conditions. Mutant p53 binds near the VEGFR2 promoter transcriptional start site and plays a role in maintaining an open conformation at that location. Relatedly, mutant p53 interacts with the SWI/SNF complex, which is required for remodeling the VEGFR2 promoter. By both querying individual genes regulated by mutant p53 and performing RNA sequencing, the results indicate that >40% of all mutant p53-regulated gene expression is mediated by SWI/SNF. We surmise that mutant p53 impacts transcription of VEGFR2 as well as myriad other genes by promoter remodeling through interaction with and likely regulation of the SWI/SNF chromatin remodeling complex. Therefore, not only might mutant p53-expressing tumors be susceptible to anti VEGF therapies, impacting SWI/SNF tumor suppressor function in mutant p53 tumors may also have therapeutic potential.


Subject(s)
Breast Neoplasms/physiopathology , Chromatin Assembly and Disassembly/genetics , Gene Expression Regulation, Neoplastic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/metabolism , HT29 Cells , Humans , MCF-7 Cells , Mutation/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Protein Conformation , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL