Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell ; 186(1): 80-97.e26, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36608661

ABSTRACT

Glucose is a universal bioenergy source; however, its role in controlling protein interactions is unappreciated, as are its actions during differentiation-associated intracellular glucose elevation. Azido-glucose click chemistry identified glucose binding to a variety of RNA binding proteins (RBPs), including the DDX21 RNA helicase, which was found to be essential for epidermal differentiation. Glucose bound the ATP-binding domain of DDX21, altering protein conformation, inhibiting helicase activity, and dissociating DDX21 dimers. Glucose elevation during differentiation was associated with DDX21 re-localization from the nucleolus to the nucleoplasm where DDX21 assembled into larger protein complexes containing RNA splicing factors. DDX21 localized to specific SCUGSDGC motif in mRNA introns in a glucose-dependent manner and promoted the splicing of key pro-differentiation genes, including GRHL3, KLF4, OVOL1, and RBPJ. These findings uncover a biochemical mechanism of action for glucose in modulating the dimerization and function of an RNA helicase essential for tissue differentiation.


Subject(s)
DEAD-box RNA Helicases , Glucose , Keratinocytes , Cell Nucleolus/metabolism , Cell Nucleus/metabolism , DEAD-box RNA Helicases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Glucose/metabolism , Keratinocytes/cytology , Keratinocytes/metabolism , Humans
2.
Curr Protoc ; 3(1): e659, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36705610

ABSTRACT

UV cross-linking-based methods are the most common tool to explore in vivo RNA-protein interactions. UV cross-linking enables the freezing of direct interactions in the cell, which can then be mapped by high-throughput sequencing through a family of methods termed CLIP-seq. CLIP-seq measures the distribution of cross-link events by purifying a protein of interest and sequencing the covalently bound RNA fragments. However, there are disagreements and ambiguities as to which proteins are RNA-binding proteins and what interactions are significant as all proteins contact all RNAs at some frequency. Here we describe a protocol for both determining RNA-protein interactions through a combination of RNA library preparation and the measurement of absolute cross-link rates, which helps determine what proteins are RNA-binding proteins and what interactions are significant. This protocol, comprising an updated form of the easyCLIP protocol, describes guidelines for RNA library preparation, oligo and protein standard construction, and the measurement of cross-link rates. These methods are easily visualizable through their fluorescent labels and can be adapted to study RNA-binding properties of both functional, high affinity RNA-binding proteins, and the accidental RNA interactions of non-RNA-binding proteins. © 2023 Wiley Periodicals LLC. Basic Protocol 1: RNA library construction Basic Protocol 2: Determining UV cross-link rates Support Protocol 1: Cross-linking and lysing cells Support Protocol 2: Adapter preparation Support Protocol 3: Preparation of cross-linked RBP standard.


Subject(s)
Chromatin Immunoprecipitation Sequencing , RNA-Binding Proteins , RNA-Binding Proteins/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , RNA/genetics , RNA/chemistry , RNA/metabolism , Gene Library , High-Throughput Nucleotide Sequencing/methods
3.
Elife ; 92020 05 07.
Article in English | MEDLINE | ID: mdl-32379046

ABSTRACT

The Xist lncRNA mediates X chromosome inactivation (XCI). Here we show that Spen, an Xist-binding repressor protein essential for XCI , binds to ancient retroviral RNA, performing a surveillance role to recruit chromatin silencing machinery to these parasitic loci. Spen loss activates a subset of endogenous retroviral (ERV) elements in mouse embryonic stem cells, with gain of chromatin accessibility, active histone modifications, and ERV RNA transcription. Spen binds directly to ERV RNAs that show structural similarity to the A-repeat of Xist, a region critical for Xist-mediated gene silencing. ERV RNA and Xist A-repeat bind the RRM domains of Spen in a competitive manner. Insertion of an ERV into an A-repeat deficient Xist rescues binding of Xist RNA to Spen and results in strictly local gene silencing in cis. These results suggest that Xist may coopt transposable element RNA-protein interactions to repurpose powerful antiviral chromatin silencing machinery for sex chromosome dosage compensation.


The genetic material inside cells is often packaged into thread-like structures called chromosomes. In humans, mice and other mammals, a pair of sex chromosomes determines the genetic or chromosomal sex of each individual. Those who inherit two "X" chromosomes are said to be chromosomally female, while chromosomal males have one "X" and one "Y" chromosome. This means females have twice as many copies of genes on the X chromosome as a male does, which turns out to be double the number that the body needs. To solve this problem, mammals have developed a strategy known as dosage compensation. The second X chromosome in females becomes "silent": its DNA remains unchanged, but none of the genes are active. A long noncoding RNA molecule called Xist is responsible for switching off the extra X genes in female cells. It does this by coating the entirety of the second X chromosome. Normally, RNA molecules transmit the coded instructions in genes to the cellular machinery that manufactures proteins. "Noncoding" RNAs like Xist, however, are RNAs that have taken on different jobs inside the cell. Researchers believe that the ancestral Xist gene may have once encoded a protein but changed over time to produce only a noncoding RNA. Carter, Xu et al. therefore set out to find out how exactly this might have happened, and also how Xist might have acquired its ability to switch genes off. Initial experiments used mouse cells grown in the laboratory, in which a protein called Spen was deleted. Spen is known to help Xist silence the X chromosome. In female cells lacking Spen, the second X chromosome remained active. Other chromosomes in male and female cells also had stretches of DNA that became active upon Spen's removal. These DNA sequences, termed endogenous retroviruses, were remnants of ancestral viral infections. In other words, Spen normally acted as an antiviral defense. Analysis of genetic sequences showed that Spen recognized endogenous retrovirus sequences resembling a key region in Xist, a region which was needed for Xist to work properly. Inserting fragments of endogenous retroviruses into a defective version of Xist lacking this region also partially restored its ability to inactivate genes, suggesting that X chromosome silencing might work by hijacking cellular defenses against viruses. That is, female cells essentially 'pretend' there is a viral infection on the second X chromosome by coating it with Xist (which mimics endogenous retroviruses), thus directing Spen to shut it down. This research is an important step towards understanding how female cells carry out dosage compensation in mammals. More broadly, it sheds new light on how ancient viruses may have shaped the evolution of noncoding RNAs in the human genome.


Subject(s)
DNA-Binding Proteins/metabolism , Endogenous Retroviruses/genetics , Mouse Embryonic Stem Cells/virology , RNA, Long Noncoding/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , X Chromosome Inactivation , X Chromosome , Animals , Binding Sites , Cell Line , DNA-Binding Proteins/genetics , Dosage Compensation, Genetic , Endogenous Retroviruses/metabolism , Female , Host-Pathogen Interactions , Mice , Mouse Embryonic Stem Cells/metabolism , Protein Binding , RNA, Long Noncoding/genetics , RNA, Viral/genetics , RNA-Binding Proteins/genetics
4.
Cell ; 176(1-2): 361-376.e17, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30580963

ABSTRACT

Here, we present Perturb-ATAC, a method that combines multiplexed CRISPR interference or knockout with genome-wide chromatin accessibility profiling in single cells based on the simultaneous detection of CRISPR guide RNAs and open chromatin sites by assay of transposase-accessible chromatin with sequencing (ATAC-seq). We applied Perturb-ATAC to transcription factors (TFs), chromatin-modifying factors, and noncoding RNAs (ncRNAs) in ∼4,300 single cells, encompassing more than 63 genotype-phenotype relationships. Perturb-ATAC in human B lymphocytes uncovered regulators of chromatin accessibility, TF occupancy, and nucleosome positioning and identified a hierarchy of TFs that govern B cell state, variation, and disease-associated cis-regulatory elements. Perturb-ATAC in primary human epidermal cells revealed three sequential modules of cis-elements that specify keratinocyte fate. Combinatorial deletion of all pairs of these TFs uncovered their epistatic relationships and highlighted genomic co-localization as a basis for synergistic interactions. Thus, Perturb-ATAC is a powerful strategy to dissect gene regulatory networks in development and disease.


Subject(s)
Epigenomics/methods , Gene Regulatory Networks/genetics , Single-Cell Analysis/methods , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Gene Regulatory Networks/physiology , High-Throughput Nucleotide Sequencing/methods , Humans , Sequence Analysis, DNA/methods , Transcription Factors/metabolism
5.
Nat Methods ; 15(3): 207-212, 2018 03.
Article in English | MEDLINE | ID: mdl-29400715

ABSTRACT

RNA-protein interactions play numerous roles in cellular function and disease. Here we describe RNA-protein interaction detection (RaPID), which uses proximity-dependent protein labeling, based on the BirA* biotin ligase, to rapidly identify the proteins that bind RNA sequences of interest in living cells. RaPID displays utility in multiple applications, including in evaluating protein binding to mutant RNA motifs in human genetic disorders, in uncovering potential post-transcriptional networks in breast cancer, and in discovering essential host proteins that interact with Zika virus RNA. To improve the BirA*-labeling component of RaPID, moreover, a new mutant BirA* was engineered from Bacillus subtilis, termed BASU, that enables >1,000-fold faster kinetics and >30-fold increased signal-to-noise ratio over the prior standard Escherichia coli BirA*, thereby enabling direct study of RNA-protein interactions in living cells on a timescale as short as 1 min.


Subject(s)
Biotin/chemistry , RNA-Binding Proteins/metabolism , RNA/metabolism , Viral Proteins/metabolism , Zika Virus/metabolism , Bacillus subtilis/metabolism , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , HEK293 Cells , Humans , Neurons/cytology , Neurons/metabolism , RNA/chemistry , RNA/genetics , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics , Zika Virus/genetics
6.
Dev Cell ; 43(2): 227-239.e5, 2017 10 23.
Article in English | MEDLINE | ID: mdl-28943242

ABSTRACT

Somatic progenitors sustain tissue self-renewal while suppressing premature differentiation. Protein arginine methyltransferases (PRMTs) affect many processes; however, their role in progenitor function is incompletely understood. PRMT1 was found to be the most highly expressed PRMT in epidermal progenitors and the most downregulated PRMT during differentiation. In targeted mouse knockouts and in long-term regenerated human mosaic epidermis in vivo, epidermal PRMT1 loss abolished progenitor self-renewal and led to premature differentiation. Mass spectrometry of the PRMT1 protein interactome identified the CSNK1a1 kinase, which also proved essential for progenitor maintenance. CSNK1a1 directly bound and phosphorylated PRMT1 to control its genomic targeting to PRMT1-sustained proliferation genes as well as PRMT1-suppressed differentiation genes. Among the latter were GRHL3, whose derepression was required for the premature differentiation seen with PRMT1 and CSNK1a1 loss. Maintenance of the progenitors thus requires cooperation by PRMT1 and CSNK1a1 to sustain proliferation gene expression and suppress premature differentiation driven by GRHL3.


Subject(s)
Casein Kinase Ialpha/metabolism , Cell Self Renewal/physiology , Epidermal Cells , Keratinocytes/cytology , Protein-Arginine N-Methyltransferases/physiology , Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Epidermis/metabolism , Humans , Infant, Newborn , Keratinocytes/metabolism , Mice , Mice, Knockout , Phosphorylation , Stem Cells/metabolism
7.
Nat Methods ; 13(6): 489-92, 2016 06.
Article in English | MEDLINE | ID: mdl-27111506

ABSTRACT

The complexity of transcriptome-wide protein-RNA interaction networks is incompletely understood. While emerging studies are greatly expanding the known universe of RNA-binding proteins, methods for the discovery and characterization of protein-RNA interactions remain resource intensive and technically challenging. Here we introduce a UV-C crosslinking and immunoprecipitation platform, irCLIP, which provides an ultraefficient, fast, and nonisotopic method for the detection of protein-RNA interactions using far less material than standard protocols.


Subject(s)
Immunoprecipitation/methods , RNA-Binding Proteins/analysis , Ultraviolet Rays , Binding Sites , Cross-Linking Reagents/chemistry , DNA, Complementary/genetics , HeLa Cells , High-Throughput Nucleotide Sequencing , Humans , Photochemical Processes , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/radiation effects , Sensitivity and Specificity , Transcriptome
8.
Nat Genet ; 48(1): 53-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26595770

ABSTRACT

Small nucleolar RNAs (snoRNAs) are conserved noncoding RNAs best studied as ribonucleoprotein (RNP) guides in RNA modification. To explore their role in cancer, we compared 5,473 tumor-normal genome pairs to identify snoRNAs with frequent copy number loss. The SNORD50A-SNORD50B snoRNA locus was deleted in 10-40% of 12 common cancers, where its loss was associated with reduced survival. A human protein microarray screen identified direct SNORD50A and SNORD50B RNA binding to K-Ras. Loss of SNORD50A and SNORD50B increased the amount of GTP-bound, active K-Ras and hyperactivated Ras-ERK1/ERK2 signaling. Loss of these snoRNAs also increased binding by farnesyltransferase to K-Ras and increased K-Ras prenylation, suggesting that KRAS mutation might synergize with SNORD50A and SNORD50B loss in cancer. In agreement with this hypothesis, CRISPR-mediated deletion of SNORD50A and SNORD50B in KRAS-mutant tumor cells enhanced tumorigenesis, and SNORD50A and SNORD50B deletion and oncogenic KRAS mutation co-occurred significantly in multiple human tumor types. SNORD50A and SNORD50B snoRNAs thus directly bind and inhibit K-Ras and are recurrently deleted in human cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , ras Proteins/metabolism , Animals , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Female , Gene Deletion , Guanosine Triphosphate/metabolism , Humans , Mice, Inbred NOD , Mutation , Neoplasms/mortality , Prenylation , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Xenograft Model Antitumor Assays , ras Proteins/genetics
9.
Dev Cell ; 35(4): 444-57, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26609959

ABSTRACT

Current gene expression network approaches commonly focus on transcription factors (TFs), biasing network-based discovery efforts away from potentially important non-TF proteins. We developed proximity analysis, a network reconstruction method that uses topological constraints of scale-free, small-world biological networks to reconstruct relationships in eukaryotic systems, independent of subcellular localization. Proximity analysis identified MPZL3 as a highly connected hub that is strongly induced during epidermal differentiation. MPZL3 was essential for normal differentiation, acting downstream of p63, ZNF750, KLF4, and RCOR1, each of which bound near the MPZL3 gene and controlled its expression. MPZL3 protein localized to mitochondria, where it interacted with FDXR, which was itself also found to be essential for differentiation. Together, MPZL3 and FDXR increased reactive oxygen species (ROS) to drive epidermal differentiation. ROS-induced differentiation is dependent upon promotion of FDXR enzymatic activity by MPZL3. ROS induction by the MPZL3 and FDXR mitochondrial proteins is therefore essential for epidermal differentiation.


Subject(s)
Cell Differentiation , Epidermal Cells , Ferredoxin-NADP Reductase/metabolism , Gene Regulatory Networks , Keratinocytes/cytology , Membrane Proteins/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Cells, Cultured , Epidermis/metabolism , Ferredoxin-NADP Reductase/genetics , Ferredoxins/metabolism , Gene Expression Regulation , Humans , Keratinocytes/metabolism , Kruppel-Like Factor 4 , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Metabolomics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , RNA, Small Interfering/genetics , Transcription Factors/metabolism
10.
Dev Cell ; 32(6): 693-706, 2015 Mar 23.
Article in English | MEDLINE | ID: mdl-25805135

ABSTRACT

Progenitor differentiation requires remodeling of genomic expression; however, in many tissues, such as epidermis, the spectrum of remodeled genes and the transcription factors (TFs) that control them are not fully defined. We performed kinetic transcriptome analysis during regeneration of differentiated epidermis and identified gene sets enriched in progenitors (594 genes), in early (159 genes), and in late differentiation (387 genes). Module mapping of 1,046 TFs identified MAF and MAFB as necessary and sufficient for progenitor differentiation. MAF:MAFB regulated 393 genes altered in this setting. Integrative analysis identified ANCR and TINCR lncRNAs as essential upstream MAF:MAFB regulators. ChIP-seq analysis demonstrated MAF:MAFB binding to known epidermal differentiation TF genes whose expression they controlled, including GRHL3, ZNF750, KLF4, and PRDM1. Each of these TFs rescued expression of specific MAF:MAFB target gene subsets in the setting of MAF:MAFB loss, indicating they act downstream of MAF:MAFB. A lncRNA-TF network is thus essential for epidermal differentiation.


Subject(s)
Cell Differentiation/genetics , Epidermal Cells , MafB Transcription Factor/genetics , Proto-Oncogene Proteins c-maf/genetics , RNA, Long Noncoding/genetics , Animals , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Transfer Techniques , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Organogenesis/genetics , Positive Regulatory Domain I-Binding Factor 1 , RNA Interference , RNA, Small Interfering , Repressor Proteins/biosynthesis , Transcription Factors/biosynthesis , Tumor Suppressor Proteins
11.
Trends Genet ; 29(1): 31-40, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23141808

ABSTRACT

More than 100 human genetic skin diseases, impacting over 20% of the population, are characterized by disrupted epidermal differentiation. A significant proportion of the 90 genes identified in these disorders to date are concentrated within several functional pathways, suggesting the emergence of organizing themes in epidermal differentiation. Among these are the Notch, transforming growth factor ß (TGFß), IκB kinase (IKK), Ras/mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K), p63, and Wnt signaling pathways, as well as core biological processes mediating calcium homeostasis, tissue integrity, cornification, and lipid biogenesis. Here, we review recent results supporting the central role of these pathways in epidermal differentiation, highlighting the integration of genetic information with functional studies to illuminate the biological actions of these pathways in humans as well as to guide development of future therapeutics to correct their dysfunction.


Subject(s)
Cell Differentiation/genetics , Epidermis/physiology , Signal Transduction/genetics , Skin Diseases/genetics , Skin Diseases/physiopathology , Animals , Epidermis/metabolism , Gene Regulatory Networks/physiology , Humans , Models, Biological , Skin Diseases/etiology
12.
Am J Hum Genet ; 91(3): 435-43, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22922031

ABSTRACT

The basis for impaired differentiation in TP63 mutant ankyloblepharon-ectodermal dysplasia-clefting (AEC) syndrome is unknown. Human epidermis harboring AEC TP63 mutants recapitulated this impairment, along with downregulation of differentiation activators, including HOPX, GRHL3, KLF4, PRDM1, and ZNF750. Gene-set enrichment analysis indicated that disrupted expression of epidermal differentiation programs under the control of ZNF750 and KLF4 accounted for the majority of disrupted epidermal differentiation resulting from AEC mutant TP63. Chromatin immunoprecipitation (ChIP) analysis and ChIP-sequencing of TP63 binding in differentiated keratinocytes revealed ZNF750 as a direct target of wild-type and AEC mutant TP63. Restoring ZNF750 to AEC model tissue rescued activator expression and differentiation, indicating that AEC TP63-mediated ZNF750 inhibition contributes to differentiation defects in AEC. Incorporating disease-causing mutants into regenerated human tissue can thus dissect pathomechanisms and identify targets that reverse disease features.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Ectodermal Dysplasia/genetics , Eye Abnormalities/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Cell Differentiation/genetics , Epidermis/metabolism , Eyelids/abnormalities , Humans , Kruppel-Like Factor 4 , Mutation , Organ Culture Techniques/methods , Transcriptome
13.
Dev Cell ; 22(3): 669-77, 2012 Mar 13.
Article in English | MEDLINE | ID: mdl-22364861

ABSTRACT

Disrupted epidermal differentiation characterizes numerous diseases that impact >25% of the population. In a search for dominant mediators of differentiation, we defined a requirement for ZNF750 in terminal epidermal differentiation. ZNF750 controlled genes mutated in numerous human skin diseases, including FLG, LOR, LCE3B, ALOXE3, and SPINK5. ZNF750 induced progenitor differentiation via an evolutionarily conserved C2H2 zinc finger motif. The epidermal master regulator, p63, bound the ZNF750 promoter and was necessary for its induction. ZNF750 restored differentiation to p63-deficient tissue, suggesting that it acts downstream of p63. A search for functionally important ZNF750 targets via analysis of ZNF750-regulated genes identified KLF4, a transcription factor that activates late epidermal differentiation. ZNF750 binds to KLF4 at multiple sites flanking the transcriptional start site and controls its expression. ZNF750 thus directly links a tissue-specifying factor, p63, to an effector of terminal differentiation, KLF4, and represents a potential future target for disorders of this process.


Subject(s)
Cell Differentiation , Epidermal Cells , Kruppel-Like Transcription Factors/physiology , Membrane Proteins/physiology , Transcription Factors/physiology , Amino Acid Sequence , Cells, Cultured , Epidermis/metabolism , Filaggrin Proteins , Foreskin/physiology , Gene Expression Regulation/physiology , Humans , Keratinocytes/physiology , Kruppel-Like Factor 4 , Male , Molecular Sequence Data , Tumor Suppressor Proteins
14.
Nat Immunol ; 9(12): 1371-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18997794

ABSTRACT

Recent studies suggest that nuclear factor kappaB-inducing kinase (NIK) is suppressed through constitutive proteasome-mediated degradation regulated by TRAF2, TRAF3 and cIAP1 or cIAP2. Here we demonstrated that the degradation of NIK occurs upon assembly of a regulatory complex through TRAF3 recruitment of NIK and TRAF2 recruitment of cIAP1 and cIAP2. In contrast to TRAF2 and TRAF3, cIAP1 and cIAP2 seem to play redundant roles in the degradation of NIK, as inhibition of both cIAPs was required for noncanonical NF-kappaB activation and increased survival and proliferation of primary B lymphocytes. Furthermore, the lethality of TRAF3 deficiency in mice could be rescued by a single NIK gene, highlighting the importance of tightly regulated NIK.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Inhibitor of Apoptosis Proteins/immunology , NF-kappa B/immunology , TNF Receptor-Associated Factor 2/immunology , TNF Receptor-Associated Factor 3/immunology , Animals , B-Lymphocytes/cytology , Cell Survival , Cells, Cultured , Enzyme Activation/immunology , Immunoblotting , Immunoprecipitation , Inhibitor of Apoptosis Proteins/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Mutant Strains , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Small Interfering , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...