Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Immunol ; 213(1): 29-39, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38767437

ABSTRACT

High-dose (HD) IL-2 was the first immuno-oncology agent approved for treating advanced renal cell carcinoma and metastatic melanoma, but its use was limited because of substantial toxicities. Multiple next-generation IL-2 agents are being developed to improve tolerability. However, a knowledge gap still exists for the genomic markers that define the target pharmacology for HD IL-2 itself. In this retrospective observational study, we collected PBMC samples from 23 patients with metastatic renal cell carcinoma who were treated with HD IL-2 between 2009 and 2015. We previously reported the results of flow cytometry analyses. In this study, we report the results of our RNA-sequencing immunogenomic survey, which was performed on bulk PBMC samples from immediately before (day 1), during (day 3), and after treatment (day 5) in cycle 1 and/or cycle 2 of the first course of HD IL-2. As part of a detailed analysis of immunogenomic response to HD IL-2 treatment, we analyzed the changes in individual genes and immune gene signatures. By day 3, most lymphoid cell types had transiently decreased, whereas myeloid transcripts increased. Although most genes and/or signatures generally returned to pretreatment expression levels by day 5, certain ones representative of B cell, NK cell, and T cell proliferation and effector functions continued to increase, along with B cell (but not T cell) oligoclonal expansion. Regulatory T cells progressively expanded during and after treatment. They showed strong negative correlation with myeloid effector cells. This detailed RNA-sequencing immunogenomic survey of IL-2 pharmacology complements results of prior flow cytometry analyses. These data provide valuable pharmacological context for assessing PBMC gene expression data from patients dosed with IL-2-related compounds that are currently in development.


Subject(s)
Carcinoma, Renal Cell , Immunotherapy , Interleukin-2 , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/genetics , Interleukin-2/administration & dosage , Interleukin-2/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Middle Aged , Female , Immunotherapy/methods , Aged , Retrospective Studies , Adult , Leukocytes, Mononuclear/immunology , Neoplasm Metastasis
2.
Clin Cancer Res ; 29(16): 3203-3213, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37233991

ABSTRACT

PURPOSE: The Piedmont study is a prospectively designed retrospective evaluation of a new 48-gene antifolate response signature (AF-PRS) in patients with locally advanced/metastatic nonsquamous (NS) non-small cell lung cancer (NSCLC) treated with pemetrexed-containing platinum doublet chemotherapy (PMX-PDC). The study tested the hypothesis that AF-PRS identifies patients with NS-NSCLC who have a higher likelihood of responding positively to PMX-PDC. The goal was to gather clinical evidence supporting AF-PRS as a potential diagnostic test. EXPERIMENTAL DESIGN: Residual pretreatment FFPE tumor samples and clinical data were analyzed from 105 patients treated with first-line (1L) PMX-PDC. Ninety-five patients had sufficient RNA sequencing (RNA-seq) data quality and clinical annotation for inclusion in the analysis. Associations between AF-PRS status and associate genes and outcome measures including progression-free survival (PFS) and clinical response were evaluated. RESULTS: Overall, 53% of patients were AF-PRS(+), which was associated with extended PFS, but not overall survival, versus AF-PRS(-) (16.6 months vs. 6.6 months; P = 0.025). In patients who were stage I to III patients at the time of treatment, PFS was further extended in AF-PRS(+) versus AF-PRS(-) (36.2 months vs. 9.3 months; P = 0.03). Complete response (CR) to therapy was noted in 14 of 95 patients. AF-PRS(+) preferentially selected a majority (79%) of CRs, which were evenly split between patients stage I to III (six of seven) and stage IV (five of seven) at the time of treatment. CONCLUSIONS: AF-PRS identified a significant population of patients with extended PFS and/or clinical response following PMX-PDC treatment. AF-PRS may be a useful diagnostic test for patients indicated for systemic chemotherapy, especially when determining the optimal PDC regimen for locally advanced disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Folic Acid Antagonists , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Pemetrexed , Platinum/therapeutic use , Folic Acid Antagonists/therapeutic use , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/adverse effects
3.
Genetics ; 222(4)2022 11 30.
Article in English | MEDLINE | ID: mdl-36218393

ABSTRACT

Transcription factors play important roles in the development of the intestinal epithelium and its ability to respond to endocrine, nutritional, and microbial signals. Hepatocyte nuclear factor 4 family nuclear receptors are liganded transcription factors that are critical for the development and function of multiple digestive organs in vertebrates, including the intestinal epithelium. Zebrafish have 3 hepatocyte nuclear factor 4 homologs, of which, hnf4a was previously shown to mediate intestinal responses to microbiota in zebrafish larvae. To discern the functions of other hepatocyte nuclear factor 4 family members in zebrafish development and intestinal function, we created and characterized mutations in hnf4g and hnf4b. We addressed the possibility of genetic redundancy amongst these factors by creating double and triple mutants which showed different rates of survival, including apparent early lethality in hnf4a; hnf4b double mutants and triple mutants. RNA sequencing performed on digestive tracts from single and double mutant larvae revealed extensive changes in intestinal gene expression in hnf4a mutants that were amplified in hnf4a; hnf4g mutants, but limited in hnf4g mutants. Changes in hnf4a and hnf4a; hnf4g mutants were reminiscent of those seen in mice including decreased expression of genes involved in intestinal function and increased expression of cell proliferation genes, and were validated using transgenic reporters and EdU labeling in the intestinal epithelium. Gnotobiotics combined with RNA sequencing also showed hnf4g has subtler roles than hnf4a in host responses to microbiota. Overall, phenotypic changes in hnf4a single mutants were strongly enhanced in hnf4a; hnf4g double mutants, suggesting a conserved partial genetic redundancy between hnf4a and hnf4g in the vertebrate intestine.


Subject(s)
Hepatocyte Nuclear Factor 4 , Intestinal Mucosa , Zebrafish Proteins , Zebrafish , Animals , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/physiology , Intestinal Mucosa/embryology , Intestinal Mucosa/metabolism , Intestines/embryology , Intestines/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
4.
Cell Mol Gastroenterol Hepatol ; 14(2): 465-493, 2022.
Article in English | MEDLINE | ID: mdl-35533983

ABSTRACT

BACKGROUND & AIMS: The intestine constantly interprets and adapts to complex combinations of dietary and microbial stimuli. However, the transcriptional strategies by which the intestinal epithelium integrates these coincident sources of information remain unresolved. We recently found that microbiota colonization suppresses epithelial activity of hepatocyte nuclear factor 4 nuclear receptor transcription factors, but their integrative regulation was unknown. METHODS: We compared adult mice reared germ-free or conventionalized with a microbiota either fed normally or after a single high-fat meal. Preparations of unsorted jejunal intestinal epithelial cells were queried using lipidomics and genome-wide assays for RNA sequencing and ChIP sequencing for the activating histone mark H3K27ac and hepatocyte nuclear factor 4 alpha. RESULTS: Analysis of lipid classes, genes, and regulatory regions identified distinct nutritional and microbial responses but also simultaneous influence of both stimuli. H3K27ac sites preferentially increased by high-fat meal in the presence of microbes neighbor lipid anabolism and proliferation genes, were previously identified intestinal stem cell regulatory regions, and were not hepatocyte nuclear factor 4 alpha targets. In contrast, H3K27ac sites preferentially increased by high-fat meal in the absence of microbes neighbor targets of the energy homeostasis regulator peroxisome proliferator activated receptor alpha, neighbored fatty acid oxidation genes, were previously identified enterocyte regulatory regions, and were hepatocyte factor 4 alpha bound. CONCLUSIONS: Hepatocyte factor 4 alpha supports a differentiated enterocyte and fatty acid oxidation program in germ-free mice, and that suppression of hepatocyte factor 4 alpha by the combination of microbes and high-fat meal may result in preferential activation of intestinal epithelial cell proliferation programs. This identifies potential transcriptional mechanisms for intestinal adaptation to multiple signals and how microbiota may modulate intestinal lipid absorption, epithelial cell renewal, and systemic energy balance.


Subject(s)
Duodenum , Gastrointestinal Microbiome , Intestinal Mucosa , Animals , Duodenum/metabolism , Duodenum/microbiology , Fatty Acids/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lipids , Mice
5.
Nat Rev Gastroenterol Hepatol ; 18(1): 7-23, 2021 01.
Article in English | MEDLINE | ID: mdl-33024279

ABSTRACT

The intestinal epithelium serves the unique and critical function of harvesting dietary nutrients, while simultaneously acting as a cellular barrier separating tissues from the luminal environment and gut microbial ecosystem. Two salient features of the intestinal epithelium enable it to perform these complex functions. First, cells within the intestinal epithelium achieve a wide range of specialized identities, including different cell types and distinct anterior-posterior patterning along the intestine. Second, intestinal epithelial cells are sensitive and responsive to the dynamic milieu of dietary nutrients, xenobiotics and microorganisms encountered in the intestinal luminal environment. These diverse identities and responsiveness of intestinal epithelial cells are achieved in part through the differential transcription of genes encoded in their shared genome. Here, we review insights from mice and other vertebrate models into the transcriptional regulatory mechanisms underlying intestinal epithelial identity and microbial responsiveness, including DNA methylation, chromatin accessibility, histone modifications and transcription factors. These studies are revealing that most transcription factors involved in intestinal epithelial identity also respond to changes in the microbiota, raising both opportunities and challenges to discern the underlying integrative transcriptional regulatory networks.


Subject(s)
Cell Differentiation/genetics , Cellular Microenvironment/physiology , Gastrointestinal Microbiome , Intestinal Mucosa/physiology , Animals , Cell Differentiation/physiology , Cellular Microenvironment/genetics , Gastrointestinal Microbiome/genetics , Gastrointestinal Microbiome/physiology , Gene Expression Regulation , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/microbiology , Intestines/microbiology , Intestines/physiology , Mice , Models, Animal , Nutrigenomics , Nutritional Physiological Phenomena/genetics , Nutritional Physiological Phenomena/physiology , Transcription, Genetic/genetics , Transcription, Genetic/physiology , Zebrafish
6.
Chem Senses ; 44(8): 615-630, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31403159

ABSTRACT

Sensory systems such as the olfactory system detect chemical stimuli and thereby determine the relationships between the animal and its surroundings. Olfaction is one of the most conserved and ancient sensory systems in vertebrates. The vertebrate olfactory epithelium is colonized by complex microbial communities, but microbial contribution to host olfactory gene expression remains unknown. In this study, we show that colonization of germ-free zebrafish and mice with microbiota leads to widespread transcriptional responses in olfactory organs as measured in bulk tissue transcriptomics and RT-qPCR. Germ-free zebrafish olfactory epithelium showed defects in pseudostratification; however, the size of the olfactory pit and the length of the cilia were not different from that of colonized zebrafish. One of the mechanisms by which microbiota control host transcriptional programs is by differential expression and activity of specific transcription factors (TFs). REST (RE1 silencing transcription factor, also called NRSF) is a zinc finger TF that binds to the conserved motif repressor element 1 found in the promoter regions of many neuronal genes with functions in neuronal development and differentiation. Colonized zebrafish and mice showed increased nasal expression of REST, and genes with reduced expression in colonized animals were strongly enriched in REST-binding motifs. Nasal commensal bacteria promoted in vitro differentiation of Odora cells by regulating the kinetics of REST expression. REST knockdown resulted in decreased Odora cell differentiation in vitro. Our results identify a conserved mechanism by which microbiota regulate vertebrate olfactory transcriptional programs and reveal a new role for REST in sensory organs.


Subject(s)
Microbiota/physiology , Nerve Tissue Proteins/genetics , Olfactory Mucosa/metabolism , Olfactory Receptor Neurons/metabolism , Repressor Proteins/genetics , Smell/genetics , Animals , Cell Line , Conserved Sequence , Gene Expression Profiling , Gene Expression Regulation , Germ-Free Life , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Olfactory Mucosa/cytology , Olfactory Mucosa/microbiology , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/microbiology , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Repressor Proteins/metabolism , Symbiosis/physiology , Zebrafish
7.
Nutrition ; 59: 29-36, 2019 03.
Article in English | MEDLINE | ID: mdl-30415160

ABSTRACT

Recent medical history has largely viewed our bacterial symbionts as pathogens to be eradicated rather than as essential partners in optimal health. However, one of the most exciting scientific advances in recent years has been the realization that commensal microorganisms (our microbiome) play vital roles in human physiology in nutrition, vitamin synthesis, drug metabolism, protection against infection, and recovery from illness. Recent data show that loss of "health-promoting" microbes and overgrowth of pathogenic bacteria (dysbiosis) in patients in the intensive care unit (ICU) appears to contribute to nosocomial infections, sepsis, and poor outcomes. Dysbiosis results from many factors, including ubiquitous antibiotic use and altered nutrition delivery in illness. Despite modern antibiotic therapy, infections and mortality from often multidrug-resistant organisms are increasing. This raises the question of whether restoration of a healthy microbiome via probiotics or synbiotics (probiotic and prebiotic combinations) to intervene on ubiquitous ICU dysbiosis would be an optimal intervention in critical illness to prevent infection and to improve recovery. This review will discuss recent innovative experimental data illuminating mechanistic pathways by which probiotics and synbiotics may provide clinical benefit. Furthermore, a review of recent clinical data demonstrating that probiotics and synbiotics can reduce complications in ICU and other populations will be undertaken. Overall, growing data for probiotic and symbiotic therapy reveal a need for definitive clinical trials of these therapies, as recently performed in healthy neonates. Future studies should target administration of probiotics and synbiotics with known mechanistic benefits to improve patient outcomes. Optimally, future probiotic and symbiotic studies will be conducted using microbiome signatures to characterize actual ICU dysbiosis and determine, and perhaps even personalize, ideal probiotic and symbiotic therapies.


Subject(s)
Critical Care/methods , Critical Illness/therapy , Dysbiosis/therapy , Probiotics/therapeutic use , Synbiotics/administration & dosage , Dysbiosis/microbiology , Humans , Intensive Care Units
8.
Zebrafish ; 15(2): 202-205, 2018 04.
Article in English | MEDLINE | ID: mdl-29173090

ABSTRACT

The completion of the zebrafish genome sequence and advances in miniaturization and multiplexing were essential to the creation of techniques such as RNA-seq, ChIP-seq, and high-throughput behavioral and chemical screens. Multiplexing was also instrumental in the recent enhancement of the classic yeast one-hybrid interaction techniques to provide unprecedented discovery capabilities for protein-DNA interactions. Unfortunately its use for zebrafish research is currently hampered by the lack of an open reading frame (ORF) clone collection. As a first step toward a complete collection, we describe a small library of transcriptional regulatory proteins comprising 142 ORFs and its potential applications.


Subject(s)
Gene Regulatory Networks , Open Reading Frames , Transcription Factors/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Cloning, Molecular , Computational Biology , Gene Expression Regulation , Gene Library , Transcription Factors/metabolism , Zebrafish/metabolism , Zebrafish Proteins/metabolism
9.
Genome Res ; 27(7): 1195-1206, 2017 07.
Article in English | MEDLINE | ID: mdl-28385711

ABSTRACT

Microbiota influence diverse aspects of intestinal physiology and disease in part by controlling tissue-specific transcription of host genes. However, host genomic mechanisms mediating microbial control of intestinal gene expression are poorly understood. Hepatocyte nuclear factor 4 (HNF4) is the most ancient family of nuclear receptor transcription factors with important roles in human metabolic and inflammatory bowel diseases, but a role in host response to microbes is unknown. Using an unbiased screening strategy, we found that zebrafish Hnf4a specifically binds and activates a microbiota-suppressed intestinal epithelial transcriptional enhancer. Genetic analysis revealed that zebrafish hnf4a activates nearly half of the genes that are suppressed by microbiota, suggesting microbiota negatively regulate Hnf4a. In support, analysis of genomic architecture in mouse intestinal epithelial cells disclosed that microbiota colonization leads to activation or inactivation of hundreds of enhancers along with drastic genome-wide reduction of HNF4A and HNF4G occupancy. Interspecies meta-analysis suggested interactions between HNF4A and microbiota promote gene expression patterns associated with human inflammatory bowel diseases. These results indicate a critical and conserved role for HNF4A in maintaining intestinal homeostasis in response to microbiota.


Subject(s)
Gastrointestinal Microbiome , Gene Expression Regulation , Hepatocyte Nuclear Factor 4/biosynthesis , Inflammatory Bowel Diseases , Zebrafish Proteins/biosynthesis , Zebrafish , Animals , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Species Specificity , Zebrafish/metabolism , Zebrafish/microbiology
10.
Subcell Biochem ; 59: 271-87, 2012.
Article in English | MEDLINE | ID: mdl-22374094

ABSTRACT

An interface coordinating lipid metabolism with proteins that regulate membrane trafficking is necessary to regulate Golgi morphology and dynamics. Such an interface facilitates the membrane deformations required for vesicularization, forms platforms for protein recruitment and assembly on appropriate sites on a membrane surface and provides lipid co-factors for optimal protein activity in the proper spatio-temporally regulated manner. Importantly, Sec14 and Sec14-like proteins are a unique superfamily of proteins that sense specific aspects of lipid metabolism, employing this information to potentiate phosphoinositide production. Therefore, Sec14 and Sec14 like proteins form central conduits to integrate multiple aspects of lipid metabolism with productive phosphoinositide signaling.


Subject(s)
Carrier Proteins/metabolism , Golgi Apparatus/physiology , Lipid Metabolism , Phosphatidylinositol Phosphates/biosynthesis , Phospholipid Transfer Proteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Animals , Biological Transport , Carrier Proteins/chemistry , Carrier Proteins/genetics , Diglycerides/metabolism , Gene Expression Regulation , Humans , Models, Molecular , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/genetics , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Signal Transduction
11.
Methods Cell Biol ; 108: 249-302, 2012.
Article in English | MEDLINE | ID: mdl-22325607

ABSTRACT

The minor cellular lipid phosphoinositides represents key regulators of diverse intracellular processes such as signal transduction at membrane-cytosol interface, regulation of membrane trafficking, cytoskeleton organization, nuclear events and the permeability, and transport functions of the membrane. The heterogeneous subcellular localization of phosphoinositides and their multiple and co-operative membrane-protein recognition mechanisms contribute to a "coincidence detection code" for the membrane-cytosol interactions in eukaryotic signaling networks. Such a "coincidence detection code" relies on the fine coordination of the broader lipid metabolism and organization, and their coupling to dedicated physiological processes. The phosphatidylinositol transfer proteins (PITPs) play a key regulatory role, essentially as "coincidence detectors" or "nanoreactors" in this "signal detection code" that spatially and temporally coordinate the diverse aspects of lipid metabolome with phosphoinositide signaling to effect various cellular functions. The integral role of PITPs in the highly conserved eukaryotic signal transduction strategy is amply demonstrated by the mammalian diseases associated with the derangements in the function of these proteins, to stress response and developmental regulation in plants, to fungal dimorphism and pathogenicity, to membrane trafficking in yeast and higher eukaryotes. The study of PITPs is fundamental to understanding of how the phosphoinositide signal transduction network is regulated and integrated to the larger lipid metabolome in diverse cellular processes. To comprehend how the PITPs integrate phosphoinositide signaling to broader lipid metabolome in diverse cellular processes, it is necessary to devise methods that can correlate the biochemical properties of these non-enzymatic proteins to biologically relevant functional insights. In this chapter, we present combinatorial approaches that primarily employ genetics and structural tools to assess the functional role of PITPs in yeast, plant and mammalian systems. An elaborate discussion on the various genetic models devised for interpreting the functional role of PITPs in relation to their operational assays has been included. We also describe the structural and biophysical methods that have advanced our understanding of how these proteins operate as "nanoreactor" molecules.


Subject(s)
Phospholipid Transfer Proteins/genetics , Animals , Arabidopsis/enzymology , Arabidopsis/genetics , Arabidopsis/metabolism , Arabidopsis Proteins/biosynthesis , Arabidopsis Proteins/metabolism , Cells, Cultured , Crystallography, X-Ray , Enzyme Assays/methods , Fungal Proteins/biosynthesis , Fungal Proteins/metabolism , Genetic Complementation Test , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/metabolism , Humans , Microscopy, Fluorescence , Microsomes, Liver/chemistry , Models, Biological , Models, Molecular , Organisms, Genetically Modified , Phosphatidylinositols/chemistry , Phosphatidylinositols/isolation & purification , Phosphatidylinositols/metabolism , Phospholipid Transfer Proteins/chemistry , Phospholipid Transfer Proteins/metabolism , Protein Binding , Protein Conformation , Radioligand Assay/methods , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Signal Transduction , Staining and Labeling , Surface Plasmon Resonance , Yeasts/enzymology , Yeasts/genetics , Yeasts/metabolism
12.
J Biol Chem ; 287(9): 6539-50, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22215675

ABSTRACT

WTX is a tumor suppressor protein that is lost or mutated in up to 30% of cases of Wilms tumor. Among its known functions, WTX interacts with the ß-transducin repeat containing family of ubiquitin ligase adaptors and promotes the ubiquitination and degradation of the transcription factor ß-catenin, a key control point in the WNT/ß-catenin signaling pathway. Here, we report that WTX interacts with a second ubiquitin ligase adaptor, KEAP1, which functions to regulate the ubiquitination of the transcription factor NRF2, a key control point in the antioxidant response. Surprisingly, we find that unlike its ability to promote the ubiquitination of ß-catenin, WTX inhibits the ubiquitination of NRF2. WTX and NRF2 compete for binding to KEAP1, and thus loss of WTX leads to rapid ubiquitination and degradation of NRF2 and a reduced response to cytotoxic insult. These results expand our understanding of the molecular mechanisms of WTX and reveal a novel regulatory mechanism governing the antioxidant response.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antioxidants/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Tumor Suppressor Proteins/metabolism , Wilms Tumor/metabolism , Adaptor Proteins, Signal Transducing/genetics , Binding, Competitive/physiology , Chromosomes, Human, X/genetics , HEK293 Cells , Humans , Kelch-Like ECH-Associated Protein 1 , Phosphorylation/physiology , RNA, Small Interfering/genetics , Serine/metabolism , Transcriptional Activation/physiology , Tumor Suppressor Proteins/genetics , Ubiquitination/physiology , Wilms Tumor/genetics , beta-Transducin Repeat-Containing Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...