Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell ; 178(4): 949-963.e18, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31353221

ABSTRACT

Estrogen receptor-positive (ER+) breast cancers frequently remain dependent on ER signaling even after acquiring resistance to endocrine agents, prompting the development of optimized ER antagonists. Fulvestrant is unique among approved ER therapeutics due to its capacity for full ER antagonism, thought to be achieved through ER degradation. The clinical potential of fulvestrant is limited by poor physicochemical features, spurring attempts to generate ER degraders with improved drug-like properties. We show that optimization of ER degradation does not guarantee full ER antagonism in breast cancer cells; ER "degraders" exhibit a spectrum of transcriptional activities and anti-proliferative potential. Mechanistically, we find that fulvestrant-like antagonists suppress ER transcriptional activity not by ER elimination, but by markedly slowing the intra-nuclear mobility of ER. Increased ER turnover occurs as a consequence of ER immobilization. These findings provide proof-of-concept that small molecule perturbation of transcription factor mobility may enable therapeutic targeting of this challenging target class.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cinnamates/pharmacology , Drug Resistance, Neoplasm , Estrogen Receptor Antagonists/therapeutic use , Female , Fulvestrant/therapeutic use , HEK293 Cells , Heterografts , Humans , Indazoles/pharmacology , Ligands , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Polymorphism, Single Nucleotide , Proteolysis/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects
2.
Proc Natl Acad Sci U S A ; 116(33): 16420-16429, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31371506

ABSTRACT

Multiple myeloma (MM) arises from malignant immunoglobulin (Ig)-secreting plasma cells and remains an incurable, often lethal disease despite therapeutic advances. The unfolded-protein response sensor IRE1α supports protein secretion by deploying a kinase-endoribonuclease module to activate the transcription factor XBP1s. MM cells may co-opt the IRE1α-XBP1s pathway; however, the validity of IRE1α as a potential MM therapeutic target is controversial. Genetic disruption of IRE1α or XBP1s, or pharmacologic IRE1α kinase inhibition, attenuated subcutaneous or orthometastatic growth of MM tumors in mice and augmented efficacy of two established frontline antimyeloma agents, bortezomib and lenalidomide. Mechanistically, IRE1α perturbation inhibited expression of key components of the endoplasmic reticulum-associated degradation machinery, as well as secretion of Ig light chains and of cytokines and chemokines known to promote MM growth. Selective IRE1α kinase inhibition reduced viability of CD138+ plasma cells while sparing CD138- cells derived from bone marrows of newly diagnosed or posttreatment-relapsed MM patients, in both US- and European Union-based cohorts. Effective IRE1α inhibition preserved glucose-induced insulin secretion by pancreatic microislets and viability of primary hepatocytes in vitro, as well as normal tissue homeostasis in mice. These results establish a strong rationale for developing kinase-directed inhibitors of IRE1α for MM therapy.


Subject(s)
Endoribonucleases/genetics , Multiple Myeloma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Aged , Animals , Bortezomib/pharmacology , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/antagonists & inhibitors , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lenalidomide/pharmacology , Male , Mice , Middle Aged , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Unfolded Protein Response/genetics , X-Box Binding Protein 1/genetics , Xenograft Model Antitumor Assays
3.
J Am Soc Nephrol ; 31(9): 2065-2082, 2020 09.
Article in English | MEDLINE | ID: mdl-32764138

ABSTRACT

BACKGROUND: Circulating APOL1 lyses trypanosomes, protecting against human sleeping sickness. Two common African gene variants of APOL1, G1 and G2, protect against infection by species of trypanosomes that resist wild-type APOL1. At the same time, the protection predisposes humans to CKD, an elegant example of balanced polymorphism. However, the exact mechanism of APOL1-mediated podocyte damage is not clear, including APOL1's subcellular localization, topology, and whether the damage is related to trypanolysis. METHODS: APOL1 topology in serum (HDL particles) and in kidney podocytes was mapped with flow cytometry, immunoprecipitation, and trypanolysis assays that tracked 170 APOL1 domain-specific monoclonal antibodies. APOL1 knockout podocytes confirmed antibody specificity. RESULTS: APOL1 localizes to the surface of podocytes, with most of the pore-forming domain (PFD) and C terminus of the Serum Resistance Associated-interacting domain (SRA-ID), but not the membrane-addressing domain (MAD), being exposed. In contrast, differential trypanolytic blocking activity reveals that the MAD is exposed in serum APOL1, with less of the PFD accessible. Low pH did not detectably alter the gross topology of APOL1, as determined by antibody accessibility, in serum or on podocytes. CONCLUSIONS: Our antibodies highlighted different conformations of native APOL1 topology in serum (HDL particles) and at the podocyte surface. Our findings support the surface ion channel model for APOL1 risk variant-mediated podocyte injury, as well as providing domain accessibility information for designing APOL1-targeted therapeutics.


Subject(s)
Apolipoprotein L1/analysis , Cell Membrane/chemistry , Podocytes/chemistry , Animals , Antibodies/immunology , Antibody Specificity , Apolipoprotein L1/blood , Apolipoprotein L1/chemistry , Apolipoprotein L1/immunology , CHO Cells , Cricetulus , Humans , Hydrogen-Ion Concentration , Podocytes/ultrastructure , Protein Domains
4.
Development ; 144(5): 866-875, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28246214

ABSTRACT

miR-1 is a small noncoding RNA molecule that modulates gene expression in heart and skeletal muscle. Loss of Drosophila miR-1 produces defects in somatic muscle and embryonic heart development, which have been partly attributed to miR-1 directly targeting Delta to decrease Notch signaling. Here, we show that overexpression of miR-1 in the fly wing can paradoxically increase Notch activity independently of its effects on Delta. Analyses of potential miR-1 targets revealed that miR-1 directly regulates the 3'UTR of the E3 ubiquitin ligase Nedd4 Analysis of embryonic and adult fly heart revealed that the Nedd4 protein regulates heart development in Drosophila Larval fly hearts overexpressing miR-1 have profound defects in actin filament organization that are partially rescued by concurrent overexpression of Nedd4. These results indicate that miR-1 and Nedd4 act together in the formation and actin-dependent patterning of the fly heart. Importantly, we have found that the biochemical and genetic relationship between miR-1 and the mammalian ortholog Nedd4-like (Nedd4l) is evolutionarily conserved in the mammalian heart, potentially indicating a role for Nedd4L in mammalian postnatal maturation. Thus, miR-1-mediated regulation of Nedd4/Nedd4L expression may serve to broadly modulate the trafficking or degradation of Nedd4/Nedd4L substrates in the heart.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , MicroRNAs/metabolism , Ubiquitin-Protein Ligases/metabolism , 3' Untranslated Regions , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Body Patterning , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Green Fluorescent Proteins/metabolism , Heart/physiology , Nedd4 Ubiquitin Protein Ligases , Phenotype , Phosphorylation , Protein Transport , Receptors, Notch/metabolism , Signal Transduction , Ubiquitination , Wings, Animal/metabolism
5.
Biotechnol Bioeng ; 114(3): 632-644, 2017 03.
Article in English | MEDLINE | ID: mdl-27666939

ABSTRACT

During antibody dependent cell cytotoxicity (ADCC) the target cells are killed by monocytes and natural killer cells. ADCC is enhanced when the antibody heavy chain's core N-linked glycan lacks the fucose molecule(s). Several strategies have been utilized to generate fully afucosylated antibodies. A commonly used and efficient approach has been knocking out the FUT8 gene of the Chinese hamster ovary (CHO) host cells, which results in expression of antibody molecules with fully afucosylated glycans. However, a major drawback of the FUT8-KO host is the requirement for undertaking two separate cell line development (CLD) efforts in order to obtain both primarily fucosylated and fully afucosylated antibody species for comparative studies in vitro and in vivo. Even more challenging is obtaining primarily fucosylated and FUT8-KO clones with similar enough product quality attributes to ensure that any observed ADCC advantage(s) can be strictly attributed to afucosylation. Here, we report generation and use of a FX knockout (FXKO) CHO host cell line that is capable of expressing antibody molecules with either primarily fucosylated or fully afucosylated glycan profiles with otherwise similar product quality attributes, depending on addition of fucose to the cell culture media. Hence, the FXKO host not only obviates the requirement for undertaking two separate CLD efforts, but it also averts the need for screening many colonies to identify clones with comparable product qualities. Finally, FXKO clones can express antibodies with the desired ratio of primarily fucosylated to afucosylated glycans when fucose is titrated into the production media, to allow achieving intended levels of FcγRIII-binding and ADCC for an antibody. Biotechnol. Bioeng. 2017;114: 632-644. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antibodies/chemistry , Fucose/metabolism , Ketone Oxidoreductases/genetics , Protein Engineering/methods , Recombinant Proteins/chemistry , Animals , Antibodies/genetics , Antibodies/metabolism , CHO Cells , CRISPR-Cas Systems , Cricetinae , Cricetulus , Fucose/chemistry , Gene Editing , Gene Knockout Techniques , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
J Biol Chem ; 290(22): 13736-48, 2015 May 29.
Article in English | MEDLINE | ID: mdl-25873391

ABSTRACT

The presence of a small number of infected but transcriptionally dormant cells currently thwarts a cure for the more than 35 million individuals infected with HIV. Reactivation of these latently infected cells may result in three fates: 1) cell death due to a viral cytopathic effect, 2) cell death due to immune clearance, or 3) a retreat into latency. Uncovering the dynamics of HIV gene expression and silencing in the latent reservoir will be crucial for developing an HIV-1 cure. Here we identify and characterize an intracellular circuit involving TRIM32, an HIV activator, and miR-155, a microRNA that may promote a return to latency in these transiently activated reservoir cells. Notably, we demonstrate that TRIM32, an E3 ubiquitin ligase, promotes reactivation from latency by directly modifying IκBα, leading to a novel mechanism of NF-κB induction not involving IκB kinase activation.


Subject(s)
HIV-1/physiology , MicroRNAs/metabolism , Transcription Factors/metabolism , Virus Latency , 3' Untranslated Regions , Amino Acid Motifs , Base Sequence , CD4-Positive T-Lymphocytes/virology , Cell Death , Gene Silencing , Genes, Reporter , Humans , I-kappa B Proteins/metabolism , Lentivirus/metabolism , Molecular Sequence Data , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Sequence Homology, Nucleic Acid , Tripartite Motif Proteins , Ubiquitin/chemistry , Ubiquitin-Protein Ligases/metabolism , Virus Replication
7.
J Biol Chem ; 289(20): 14263-71, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24719334

ABSTRACT

MicroRNA (miRNA) maturation is regulated by interaction of particular miRNA precursors with specific RNA-binding proteins. Following their biogenesis, mature miRNAs are incorporated into the RNA-induced silencing complex (RISC) where they interact with mRNAs to negatively regulate protein production. However, little is known about how mature miRNAs are regulated at the level of their activity. To address this, we screened for proteins differentially bound to the mature form of the miR-1 or miR-133 miRNA families. These muscle-enriched, co-transcribed miRNA pairs cooperate to suppress smooth muscle gene expression in the heart. However, they also have opposing roles, with the miR-1 family, composed of miR-1 and miR-206, promoting myogenic differentiation, whereas miR-133 maintains the progenitor state. Here, we describe a physical interaction between TDP-43, an RNA-binding protein that forms aggregates in the neuromuscular disease, amyotrophic lateral sclerosis, and the miR-1, but not miR-133, family. Deficiency of the TDP-43 Drosophila ortholog enhanced dmiR-1 activity in vivo. In mammalian cells, TDP-43 limited the activity of both miR-1 and miR-206, but not the miR-133 family, by disrupting their RISC association. Consistent with TDP-43 dampening miR-1/206 activity, protein levels of the miR-1/206 targets, IGF-1 and HDAC4, were elevated in TDP-43 transgenic mouse muscle. This occurred without corresponding Igf-1 or Hdac4 mRNA increases and despite higher miR-1 and miR-206 expression. Our findings reveal that TDP-43 negatively regulates the activity of the miR-1 family of miRNAs by limiting their bioavailability for RISC loading and suggest a processing-independent mechanism for differential regulation of miRNA activity.


Subject(s)
DNA-Binding Proteins/metabolism , MicroRNAs/metabolism , RNA-Induced Silencing Complex/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Argonaute Proteins/metabolism , Drosophila melanogaster/genetics , Humans , Male , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Protein Binding
8.
Nature ; 460(7257): 863-8, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19587682

ABSTRACT

An open chromatin largely devoid of heterochromatin is a hallmark of stem cells. It remains unknown whether an open chromatin is necessary for the differentiation potential of stem cells, and which molecules are needed to maintain open chromatin. Here we show that the chromatin remodelling factor Chd1 is required to maintain the open chromatin of pluripotent mouse embryonic stem cells. Chd1 is a euchromatin protein that associates with the promoters of active genes, and downregulation of Chd1 leads to accumulation of heterochromatin. Chd1-deficient embryonic stem cells are no longer pluripotent, because they are incapable of giving rise to primitive endoderm and have a high propensity for neural differentiation. Furthermore, Chd1 is required for efficient reprogramming of fibroblasts to the pluripotent stem cell state. Our results indicate that Chd1 is essential for open chromatin and pluripotency of embryonic stem cells, and for somatic cell reprogramming to the pluripotent state.


Subject(s)
Chromatin Assembly and Disassembly , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Euchromatin/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Biomarkers , Cell Proliferation , Cells, Cultured , Cellular Reprogramming , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Endoderm/metabolism , Euchromatin/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Histones/metabolism , Methylation , Mice , Neurogenesis , Neurons/cytology , Neurons/metabolism , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic/genetics , RNA Interference
9.
Nat Commun ; 14(1): 902, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36804928

ABSTRACT

CRISPR-mediated transcriptional activation (CRISPRa) is a powerful technology for inducing gene expression from endogenous loci with exciting applications in high throughput gain-of-function genomic screens and the engineering of cell-based models. However, current strategies for generating potent, stable, CRISPRa-competent cell lines present limitations for the broad utility of this approach. Here, we provide a high-efficiency, self-selecting CRISPRa enrichment strategy, which combined with piggyBac transposon technology enables rapid production of CRISPRa-ready cell populations compatible with a variety of downstream assays. We complement this with an optimized guide RNA scaffold that significantly enhances CRISPRa functionality. Finally, we describe a synthetic guide RNA tool set that enables transient, population-wide gene activation when used with the self-selecting CRISPRa system. Taken together, this versatile platform greatly enhances the potential for CRISPRa across a wide variety of cellular contexts.


Subject(s)
Genome , RNA , Transcriptional Activation , Cell Line , CRISPR-Cas Systems/genetics
10.
Nat Biotechnol ; 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37857725

ABSTRACT

The broad application of precision cancer immunotherapies is limited by the number of validated neoepitopes that are common among patients or tumor types. To expand the known repertoire of shared neoantigen-human leukocyte antigen (HLA) complexes, we developed a high-throughput platform that coupled an in vitro peptide-HLA binding assay with engineered cellular models expressing individual HLA alleles in combination with a concatenated transgene harboring 47 common cancer neoantigens. From more than 24,000 possible neoepitope-HLA combinations, biochemical and computational assessment yielded 844 unique candidates, of which 86 were verified after immunoprecipitation mass spectrometry analyses of engineered, monoallelic cell lines. To evaluate the potential for immunogenicity, we identified T cell receptors that recognized select neoepitope-HLA pairs and elicited a response after introduction into human T cells. These cellular systems and our data on therapeutically relevant neoepitopes in their HLA contexts will aid researchers studying antigen processing as well as neoepitope targeting therapies.

11.
Nat Commun ; 13(1): 4739, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35961968

ABSTRACT

CRISPR technologies have advanced cancer modelling in mice, but CRISPR activation (CRISPRa) methods have not been exploited in this context. We establish a CRISPRa mouse (dCas9a-SAMKI) for inducing gene expression in vivo and in vitro. Using dCas9a-SAMKI primary lymphocytes, we induce B cell restricted genes in T cells and vice versa, demonstrating the power of this system. There are limited models of aggressive double hit lymphoma. Therefore, we transactivate pro-survival BCL-2 in Eµ-MycT/+;dCas9a-SAMKI/+ haematopoietic stem and progenitor cells. Mice transplanted with these cells rapidly develop lymphomas expressing high BCL-2 and MYC. Unlike standard Eµ-Myc lymphomas, BCL-2 expressing lymphomas are highly sensitive to the BCL-2 inhibitor venetoclax. We perform genome-wide activation screens in these lymphoma cells and find a dominant role for the BCL-2 protein A1 in venetoclax resistance. Here we show the potential of our CRISPRa model for mimicking disease and providing insights into resistance mechanisms towards targeted therapies.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Lymphoma , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Lymphoma/drug therapy , Lymphoma/genetics , Lymphoma/pathology , Mice , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Sulfonamides
12.
J Biol Chem ; 284(27): 18515-24, 2009 Jul 03.
Article in English | MEDLINE | ID: mdl-19419954

ABSTRACT

The cell surface receptor tyrosine kinase HER2/neu enhances tumor metastasis. Recent studies suggest that deregulated microRNA (miRNA) expression promotes invasion and metastasis of cancer cells; we therefore explored the possibility that HER2/neu signaling induces the expression of specific miRNAs involved in this process. We identified a putative oncogenic miRNA, miR-21, whose expression is correlated with HER2/neu up-regulation and is functionally involved in HER2/neu-induced cell invasion. We show that miR-21 is up-regulated via the MAPK (ERK1/2) pathway upon stimulation of HER2/neu signaling in breast cancer cells, and overexpression of other ERK1/2 activators such as RASV12 or ID-1 is sufficient to induce miR-21 up-regulation in HER2/neu-negative breast cancer cells. Furthermore, the metastasis suppressor protein PDCD4 (programmed cell death 4) is down-regulated by miR-21 in breast cancer cells expressing HER2/neu. Our data reveal a mechanism for HER2/neu-induced cancer cell invasion via miRNA deregulation. In addition, our results identify miR-21 as a potential therapeutic target for the prevention of breast cancer invasion and metastasis.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , MAP Kinase Signaling System/genetics , MicroRNAs/metabolism , Receptor, ErbB-2/genetics , Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , MAP Kinase Kinase 1/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , RNA-Binding Proteins/genetics , Receptor, ErbB-2/metabolism , Up-Regulation/genetics
14.
Biotechnol Prog ; 36(3): e2951, 2020 05.
Article in English | MEDLINE | ID: mdl-31846224

ABSTRACT

Chinese hamster ovary (CHO) cells have been adapted to grow in serum-free media and in suspension culture to facilitate manufacturing needs. Some CHO cell lines, however, tend to form cell aggregates while being cultured in suspension. This can result in reduced viability and capacity for single cell cloning (SCC) via limiting dilution, and process steps to mitigate cell aggregate formation, for example, addition of anti-cell-aggregation agents. In this study, we have identified endothelial intercellular cell adhesion molecule 1 (ICAM-1) as a key protein promoting cell aggregate formation in a production competent CHO cell line, which is prone to cell aggregate formation. Knocking out (KO) the ICAM-1 gene significantly decreased cell aggregate formation in the culture media without anti-cell-aggregation reagent. This trait can simplify the process of transfection, selection, automated clone isolation, and so on. Evaluation in standard cell line development of ICAM-1 KO and wild-type CHO hosts did not reveal any noticeable impacts on titer or product quality. Furthermore, analysis of a derived nonaggregating cell line showed significant reductions in expression of cell adhesion proteins. Overall, our data suggest that deletion of ICAM-1 and perhaps other cell adhesion proteins can reduce cell aggregate formation and improve clonality assurance during SCC.


Subject(s)
Cell Adhesion/drug effects , Cell Aggregation/genetics , Intercellular Adhesion Molecule-1/genetics , Animals , CHO Cells/drug effects , Clone Cells/drug effects , Cricetinae , Cricetulus , Culture Media, Serum-Free/pharmacology , Gene Expression Regulation/genetics , Gene Knockout Techniques , Humans
15.
Nat Struct Mol Biol ; 27(2): 210-220, 2020 02.
Article in English | MEDLINE | ID: mdl-32015554

ABSTRACT

Neddylation is the post-translational protein modification most closely related to ubiquitination. Whereas the ubiquitin-like protein NEDD8 is well studied for its role in activating cullin-RING E3 ubiquitin ligases, little is known about other substrates. We developed serial NEDD8-ubiquitin substrate profiling (sNUSP), a method that employs NEDD8 R74K knock-in HEK293 cells, allowing discrimination of endogenous NEDD8- and ubiquitin-modification sites by MS after Lys-C digestion and K-εGG-peptide enrichment. Using sNUSP, we identified 607 neddylation sites dynamically regulated by the neddylation inhibitor MLN4924 and the de-neddylating enzyme NEDP1, implying that many non-cullin proteins are neddylated. Among the candidates, we characterized lysine 112 of the actin regulator cofilin as a novel neddylation event. Global inhibition of neddylation in developing neurons leads to cytoskeletal defects, altered actin dynamics and neurite growth impairments, whereas site-specific neddylation of cofilin at K112 regulates neurite outgrowth, suggesting that cofilin neddylation contributes to the regulation of neuronal actin organization.


Subject(s)
Actins/metabolism , Cofilin 1/metabolism , NEDD8 Protein/metabolism , Neurons/metabolism , Animals , Cell Line , Cells, Cultured , Gene Knock-In Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , NEDD8 Protein/genetics , Neurons/cytology , Point Mutation , Rats , Rats, Sprague-Dawley , Ubiquitin/metabolism , Ubiquitination
18.
FEBS Lett ; 580(22): 5150-4, 2006 Oct 02.
Article in English | MEDLINE | ID: mdl-16970942

ABSTRACT

The dual oxidase isozymes Duox1 and Duox2 exhibit functional NADPH:O(2) oxidoreductase activity in thyroid and respiratory tract cells and are thought to be essential for H(2)O(2) generation in these tissues. However, it is not universally accepted that the heme peroxidase domains of the Duox isozymes are functional. To address this question, we modulated Duox2 expression in human tracheobronchial epithelial (TBE) cell culture systems and quantified peroxidase activity. We discovered that interferon-gamma (IFN-gamma) induced robust peroxidase activity in TBE cells that paralleled Duox2 expression. IFN-gamma-induced peroxidase activity was abolished in the presence of sodium azide, which implicated the activation of a heme peroxidase. IFN-gamma-induced peroxidase activity was abolished in TBE cell lines expressing anti-Duox2 short hairpin RNA transcripts. Together, these data unequivocally demonstrated that Duox2 contains a functional heme peroxidase in intact respiratory tract epithelium.


Subject(s)
Flavoproteins/biosynthesis , Gene Expression Regulation, Enzymologic , NADPH Oxidases/biosynthesis , Peroxidases/biosynthesis , Respiratory Mucosa/enzymology , Antineoplastic Agents/pharmacology , Cell Line, Transformed , Dual Oxidases , Enzyme Activation/drug effects , Enzyme Activation/genetics , Flavoproteins/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Humans , Interferon-gamma/pharmacology , NADPH Oxidases/genetics , Organ Specificity , Peroxidases/genetics , RNA, Antisense/biosynthesis , RNA, Antisense/genetics , Thyroid Gland/enzymology
19.
Elife ; 2: e01323, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24252873

ABSTRACT

microRNA-1 (miR-1) is an evolutionarily conserved, striated muscle-enriched miRNA. Most mammalian genomes contain two copies of miR-1, and in mice, deletion of a single locus, miR-1-2, causes incompletely penetrant lethality and subtle cardiac defects. Here, we report that deletion of miR-1-1 resulted in a phenotype similar to that of the miR-1-2 mutant. Compound miR-1 knockout mice died uniformly before weaning due to severe cardiac dysfunction. miR-1-null cardiomyocytes had abnormal sarcomere organization and decreased phosphorylation of the regulatory myosin light chain-2 (MLC2), a critical cytoskeletal regulator. The smooth muscle-restricted inhibitor of MLC2 phosphorylation, Telokin, was ectopically expressed in the myocardium, along with other smooth muscle genes. miR-1 repressed Telokin expression through direct targeting and by repressing its transcriptional regulator, Myocardin. Our results reveal that miR-1 is required for postnatal cardiac function and reinforces the striated muscle phenotype by regulating both transcriptional and effector nodes of the smooth muscle gene expression network. DOI: http://dx.doi.org/10.7554/eLife.01323.001.


Subject(s)
Gene Expression , MicroRNAs/physiology , Muscle, Smooth/metabolism , Myocardium/metabolism , Sarcomeres , Animals , Mice , Mice, Knockout , MicroRNAs/genetics , Phosphorylation
20.
Curr Protoc Neurosci ; Chapter 5: Unit 5.26, 2007 Apr.
Article in English | MEDLINE | ID: mdl-18428658

ABSTRACT

RNA interference (RNAi) refers to the process by which 21- to 23-nucleotide short interfering RNAs (siRNAs) mediate post-transcriptional degradation of homologous mRNA transcripts. This process is carried out by an endogenous pathway that centers on the use of endogenously encoded small RNAs, and can be hijacked to knock down the expression of any target protein by introducing a specific siRNA into a cell. Stable knockdown can be obtained by constitutive expression of the siRNA from the host chromosome. Retroviruses, such as lentivirus, provide a convenient vector by which to integrate RNAi expression constructs. Lentiviruses can infect nondividing cells, thereby allowing knockdown in cells such as mature neurons. This unit provides methods to design and clone siRNAs into a lentiviral vector. Additional protocols describe production and titering of the lentivirus, as well as safety testing. Finally, methods are provided for infecting neurons in culture and in vivo with RNAi lentivirus.


Subject(s)
Lentivirus/genetics , Molecular Biology/methods , Neurons/physiology , RNA Interference , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL