Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 220
Filter
1.
Cell ; 181(7): 1502-1517.e23, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32559462

ABSTRACT

RNA viruses are a major human health threat. The life cycles of many highly pathogenic RNA viruses like influenza A virus (IAV) and Lassa virus depends on host mRNA, because viral polymerases cleave 5'-m7G-capped host transcripts to prime viral mRNA synthesis ("cap-snatching"). We hypothesized that start codons within cap-snatched host transcripts could generate chimeric human-viral mRNAs with coding potential. We report the existence of this mechanism of gene origination, which we named "start-snatching." Depending on the reading frame, start-snatching allows the translation of host and viral "untranslated regions" (UTRs) to create N-terminally extended viral proteins or entirely novel polypeptides by genetic overprinting. We show that both types of chimeric proteins are made in IAV-infected cells, generate T cell responses, and contribute to virulence. Our results indicate that during infection with IAV, and likely a multitude of other human, animal and plant viruses, a host-dependent mechanism allows the genesis of hybrid genes.


Subject(s)
RNA Caps/genetics , RNA Virus Infections/genetics , Recombinant Fusion Proteins/genetics , 5' Untranslated Regions/genetics , Animals , Cattle , Cell Line , Cricetinae , Dogs , Humans , Influenza A virus/metabolism , Mice , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Open Reading Frames/genetics , RNA Caps/metabolism , RNA Virus Infections/metabolism , RNA Viruses/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Recombinant Fusion Proteins/metabolism , Transcription, Genetic/genetics , Viral Proteins/metabolism , Virus Replication/genetics
2.
Proc Natl Acad Sci U S A ; 118(29)2021 07 20.
Article in English | MEDLINE | ID: mdl-34257153

ABSTRACT

Chimeric terpene synthases, which consist of C-terminal prenyltransferase (PT) and N-terminal class I terpene synthase (TS) domains (termed PTTSs here), is unique to fungi and produces structurally diverse di- and sesterterpenes. Prior to this study, 20 PTTSs had been functionally characterized. Our understanding of the origin and functional evolution of PTTS genes is limited. Our systematic search of sequenced fungal genomes among diverse taxa revealed that PTTS genes were restricted to Dikarya. Phylogenetic findings indicated different potential models of the origin and evolution of PTTS genes. One was that PTTS genes originated in the common Dikarya ancestor and then underwent frequent gene loss among various subsequent lineages. To understand their functional evolution, we selected 74 PTTS genes for biochemical characterization in an efficient precursor-providing yeast system employing chassis-based, robot-assisted, high-throughput automatic assembly. We found 34 PTTS genes that encoded active enzymes and collectively produced 24 di- and sesterterpenes. About half of these di- and sesterterpenes were also the products of the 20 known PTTSs, indicating functional conservation, whereas the PTTS products included the previously unknown sesterterpenes, sesterevisene (1), and sesterorbiculene (2), suggesting that a diversity of PTTS products awaits discovery. Separating functional PTTSs into two monophyletic groups implied that an early gene duplication event occurred during the evolution of the PTTS family followed by functional divergence with the characteristics of distinct cyclization mechanisms.


Subject(s)
Alkyl and Aryl Transferases/genetics , Fungal Proteins/genetics , Mutant Chimeric Proteins/genetics , Alkyl and Aryl Transferases/metabolism , Diterpenes/chemistry , Diterpenes/metabolism , Evolution, Molecular , Fungal Proteins/metabolism , Fungi/classification , Fungi/enzymology , Fungi/genetics , Genome, Fungal/genetics , Molecular Structure , Mutant Chimeric Proteins/metabolism , Mutation , Phylogeny , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sesterterpenes/chemistry , Sesterterpenes/metabolism
3.
Nucleic Acids Res ; 49(5): 2509-2521, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33555349

ABSTRACT

The paucity of recurrent mutations has hampered efforts to understand and treat neuroblastoma. Alternative splicing and splicing-dependent RNA-fusions represent mechanisms able to increase the gene product repertoire but their role in neuroblastoma remains largely unexplored. Here we investigate the presence and possible roles of aberrant splicing and splicing-dependent RNA-fusion transcripts in neuroblastoma. In addition, we attend to establish whether the spliceosome can be targeted to treat neuroblastoma. Through analysis of RNA-sequenced neuroblastoma we show that elevated expression of splicing factors is a strong predictor of poor clinical outcome. Furthermore, we identified >900 primarily intrachromosomal fusions containing canonical splicing sites. Fusions included transcripts from well-known oncogenes, were enriched for proximal genes and in chromosomal regions commonly gained or lost in neuroblastoma. As a proof-of-principle that these fusions can generate altered gene products, we characterized a ZNF451-BAG2 fusion, producing a truncated BAG2-protein which inhibited retinoic acid induced differentiation. Spliceosome inhibition impeded neuroblastoma fusion expression, induced apoptosis and inhibited xenograft tumor growth. Our findings elucidate a splicing-dependent mechanism generating altered gene products in neuroblastoma and show that the spliceosome is a potential target for clinical intervention.


Subject(s)
Molecular Chaperones/genetics , Mutant Chimeric Proteins/genetics , Neuroblastoma/genetics , RNA Splicing , Spliceosomes/drug effects , Aminoacyltransferases/metabolism , Animals , Apoptosis , Cell Differentiation , Cell Line, Tumor , Female , Gene Fusion , HSC70 Heat-Shock Proteins/metabolism , Humans , Mice, Nude , Molecular Chaperones/metabolism , Mutant Chimeric Proteins/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Sequence Deletion , Transcription Factors/metabolism , tau Proteins/metabolism
4.
Plant J ; 102(1): 116-128, 2020 04.
Article in English | MEDLINE | ID: mdl-31736145

ABSTRACT

Heterosis is the phenomenon in which hybrid progeny exhibits superior traits in comparison with those of their parents. Genomic variations between the two parental genomes may generate epistasis interactions, which is one of the genetic hypotheses explaining heterosis. We postulate that protein-protein interactions specific to F1 hybrids (F1 -specific PPIs) may occur when two parental genomes combine, as the proteome of each parent may supply novel interacting partners. To test our assumption, an inter-subspecies hybrid interactome was simulated by in silico PPI prediction between rice japonica (cultivar Nipponbare) and indica (cultivar 9311). Four-thousand, six-hundred and twelve F1 -specific PPIs accounting for 20.5% of total PPIs in the hybrid interactome were found. Genes participating in F1 -specific PPIs tend to encode metabolic enzymes and are generally localized in genomic regions harboring metabolic gene clusters. To test the genetic effect of F1 -specific PPIs in heterosis, genomic selection analysis was performed for trait prediction with additive, dominant and epistatic effects separately considered in the model. We found that the removal of single nucleotide polymorphisms associated with F1 -specific PPIs reduced prediction accuracy when epistatic effects were considered in the model, but no significant changes were observed when additive or dominant effects were considered. In summary, genomic divergence widely dispersed between japonica and indica rice may generate F1 -specific PPIs, part of which may accumulatively contribute to heterosis according to our computational analysis. These candidate F1 -specific PPIs, especially for those involved in metabolic biosynthesis pathways, are worthy of experimental validation when large-scale protein interactome datasets are generated in hybrid rice in the future.


Subject(s)
Epistasis, Genetic , Hybrid Vigor , Oryza/genetics , Plant Proteins/genetics , Protein Interaction Maps , Epistasis, Genetic/genetics , Hybrid Vigor/genetics , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Mutation, Missense , Plant Proteins/metabolism , Plant Proteins/physiology , Protein Interaction Maps/genetics
5.
J Gen Virol ; 102(9)2021 09.
Article in English | MEDLINE | ID: mdl-34494949

ABSTRACT

Transmission of the crinivirus, lettuce infectious yellows virus (LIYV), is determined by a minor coat protein (CPm)-mediated virion retention mechanism located in the foregut of its whitefly vector. To better understand the functions of LIYV CPm, chimeric CPm mutants engineered with different lengths of the LIYV CPm amino acid sequence and that of the crinivirus, lettuce chlorosis virus (LCV), were constructed based on bioinformatics and sequence alignment data. The 485 amino acid-long chimeric CPm of LIYV mutant, CPmP-1, contains 60 % (from position 3 to 294) of LCV CPm amino acids. The chimeric CPm of mutants CPmP-2, CPmP-3 and CPmP-4 contains 46 (position 3 to 208), 51 (position 3 to 238) and 41 % (position 261 to 442) of LCV CPm amino acids, respectively. All four mutants moved systemically, expressed the chimeric CPm and formed virus particles. However, following acquisition feeding of the virus preparations, only CPmP-1 was retained in the foreguts of a significant number of vectors and transmitted. In immuno-gold labelling transmission electron microscopy (IGL-TEM) analysis, CPmP-1 particles were distinctly labelled by antibodies directed against the LCV but not LIYV CPm. In contrast, CPmP-4 particles were not labelled by antibodies directed against the LCV or LIYV CPm, while CPmP-2 and -3 particles were weakly labelled by anti-LIYV CPm but not anti-LCV CPm antibodies. The unique antibody recognition and binding pattern of CPmP-1 was also displayed in the foreguts of whitefly vectors that fed on CPmP-1 virions. These results are consistent with the hypothesis that the chimeric CPm of CPmP-1 is incorporated into functional virions, with the LCV CPm region being potentially exposed on the surface and accessible to anti-LCV CPm antibodies.


Subject(s)
Capsid Proteins/metabolism , Crinivirus/physiology , Hemiptera/virology , Insect Vectors/virology , Nicotiana/virology , Plant Diseases/virology , Animals , Capsid Proteins/chemistry , Capsid Proteins/genetics , Crinivirus/genetics , Digestive System/virology , Genetic Engineering , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/metabolism , Mutation , Plants, Genetically Modified/virology , Virion/physiology
6.
Microbiology (Reading) ; 167(2)2021 02.
Article in English | MEDLINE | ID: mdl-33357282

ABSTRACT

Mycobacterium avium subspecies hominissuis (MAH) is a pathogen that causes various non-tuberculous mycobacterial diseases in humans and animals worldwide. Among the genus, MAH is characterized by relatively slow growth. Here, we isolated a rapidly growing variant of the MAH 104 strain. The variant strain (named N104) exhibited an enhanced growth rate and higher motility compared to the parent MAH 104 strain (P104). Whole-genome sequencing analysis of N104 revealed the loss of the stop codon of MAV_RS14660 due to a single nucleotide replacement, resulting in the substitution of the codon for tryptophan. Notably, exclusion of the stop codon ligated the open reading frames and caused the fusion of two adjacent proteins. A revertant parent strain, in which a mutation was introduced to restore the stop codon, revealed that elimination of the stop codon in MAV_RS14660 was responsible for the N104 phenotype. Furthermore, we analysed the phenotypes of the parent and mutated strains by determining the functions of the MAV_RS14660 and MAV_RS14655 coding regions flanking the stop codon. The mutant strains, expected to express a fusion protein, exhibited increased resistance to antimicrobial drugs and exogenous copper toxicity compared to that of the parent strains. These findings suggest that the fusion of the MAV_RS14660- and MAV_RS14655-encoding regions in the mutant N104 strain could be related to the modified functions of these intrinsic proteins.


Subject(s)
Bacterial Proteins/genetics , Mycobacterium/growth & development , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Codon, Terminator/genetics , Copper/pharmacology , Drug Resistance, Bacterial/genetics , Genome, Bacterial/genetics , Humans , Locomotion/genetics , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Mycobacterium/drug effects , Mycobacterium/genetics , Mycobacterium Infections/microbiology , Point Mutation
7.
J Virol ; 94(18)2020 08 31.
Article in English | MEDLINE | ID: mdl-32641474

ABSTRACT

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that can cause severe clinical disease in allograft recipients and infants infected in utero Virus-neutralizing antibodies defined in vitro have been proposed to confer protection against HCMV infection, and the virion envelope glycoprotein B (gB) serves as a major target of neutralizing antibodies. The viral fusion protein gB is nonfusogenic on its own and requires glycoproteins H (gH) and L (gL) for membrane fusion, which is in contrast to requirements of related class III fusion proteins, including vesicular stomatitis virus glycoprotein G (VSV-G) or baculovirus gp64. To explore requirements for gB's fusion activity, we generated a set of chimeras composed of gB and VSV-G or gp64, respectively. These gB chimeras were intrinsically fusion active and led to the formation of multinucleated cell syncytia when expressed in the absence of other viral proteins. Utilizing a panel of virus-neutralizing gB-specific monoclonal antibodies (MAbs), we could demonstrate that syncytium formation of the fusogenic gB/VSV-G chimera can be significantly inhibited by only a subset of neutralizing MAbs which target antigenic domain 5 (AD-5) of gB. This observation argues for differential modes of action of neutralizing anti-gB MAbs and suggests that blocking the membrane fusion function of gB could be one mechanism of antibody-mediated virus neutralization. In addition, our data have important implications for the further understanding of the conformation of gB that promotes membrane fusion as well as the identification of structures in AD-5 that could be targeted by antibodies to block this early step in HCMV infection.IMPORTANCE HCMV is a major global health concern, and antiviral chemotherapy remains problematic due to toxicity of available compounds and the emergence of drug-resistant viruses. Thus, an HCMV vaccine represents a priority for both governmental and pharmaceutical research programs. A major obstacle for the development of a vaccine is a lack of knowledge of the nature and specificities of protective immune responses that should be induced by such a vaccine. Glycoprotein B of HCMV is an important target for neutralizing antibodies and, hence, is often included as a component of intervention strategies. By generation of fusion-active gB chimeras, we were able to identify target structures of neutralizing antibodies that potently block gB-induced membrane fusion. This experimental system provides an approach to screen for antibodies that interfere with gB's fusogenic activity. In summary, our data will likely contribute to both rational vaccine design and the development of antibody-based therapies against HCMV.


Subject(s)
Antibodies, Neutralizing/pharmacology , Cytomegalovirus/genetics , Mutant Chimeric Proteins/genetics , Viral Envelope Proteins/genetics , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Antibodies, Viral/pharmacology , Binding Sites , Cell Fusion , Cell Line , Cytomegalovirus/drug effects , Cytomegalovirus/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/virology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression , Giant Cells/drug effects , Giant Cells/metabolism , Giant Cells/ultrastructure , Giant Cells/virology , HEK293 Cells , Humans , Mice , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/metabolism , Primary Cell Culture , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/virology , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Envelope Proteins/metabolism
8.
Nucleic Acids Res ; 47(D1): D994-D1004, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30407583

ABSTRACT

Gene fusion is one of the hallmarks of cancer genome via chromosomal rearrangement initiated by DNA double-strand breakage. To date, many fusion genes (FGs) have been established as important biomarkers and therapeutic targets in multiple cancer types. To better understand the function of FGs in cancer types and to promote the discovery of clinically relevant FGs, we built FusionGDB (Fusion Gene annotation DataBase) available at https://ccsm.uth.edu/FusionGDB. We collected 48 117 FGs across pan-cancer from three representative fusion gene resources: the improved database of chimeric transcripts and RNA-seq data (ChiTaRS 3.1), an integrative resource for cancer-associated transcript fusions (TumorFusions), and The Cancer Genome Atlas (TCGA) fusions by Gao et al. For these ∼48K FGs, we performed functional annotations including gene assessment across pan-cancer fusion genes, open reading frame (ORF) assignment, and retention search of 39 protein features based on gene structures of multiple isoforms with different breakpoints. We also provided the fusion transcript and amino acid sequences according to multiple breakpoints and transcript isoforms. Our analyses identified 331, 303 and 667 in-frame FGs with retaining kinase, DNA-binding, and epigenetic factor domains, respectively, as well as 976 FGs lost protein-protein interaction. FusionGDB provides six categories of annotations: FusionGeneSummary, FusionProtFeature, FusionGeneSequence, FusionGenePPI, RelatedDrug and RelatedDisease.


Subject(s)
Databases, Genetic , Gene Fusion , Mutant Chimeric Proteins/genetics , Neoplasms/genetics , Amino Acid Sequence , Molecular Sequence Annotation , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/metabolism , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics , Open Reading Frames , Protein Interaction Mapping , User-Computer Interface
9.
Int J Mol Sci ; 22(22)2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34830054

ABSTRACT

Chimeric RNAs are often associated with chromosomal rearrangements in cancer. In addition, they are also widely detected in normal tissues, contributing to transcriptomic complexity. Despite their prevalence, little is known about the characteristics and functions of chimeric RNAs. Here, we examine the genetic structure and biological roles of CLEC12A-MIR223HG, a novel chimeric transcript produced by the fusion of the cell surface receptor CLEC12A and the miRNA-223 host gene (MIR223HG), first identified in chronic myeloid leukemia (CML) patients. Surprisingly, we observed that CLEC12A-MIR223HG is not just expressed in CML, but also in a variety of normal tissues and cell lines. CLEC12A-MIR223HG expression is elevated in pro-monocytic cells resistant to chemotherapy and during monocyte-to-macrophage differentiation. We observed that CLEC12A-MIR223HG is a product of trans-splicing rather than a chromosomal rearrangement and that transcriptional activation of CLEC12A with the CRISPR/Cas9 Synergistic Activation Mediator (SAM) system increases CLEC12A-MIR223HG expression. CLEC12A-MIR223HG translates into a chimeric protein, which largely resembles CLEC12A but harbours an altered C-type lectin domain altering key disulphide bonds. These alterations result in differences in post-translational modifications, cellular localization, and protein-protein interactions. Taken together, our observations support a possible involvement of CLEC12A-MIR223HG in the regulation of CLEC12A function. Our workflow also serves as a template to study other uncharacterized chimeric RNAs.


Subject(s)
Gene Fusion , Lectins, C-Type/genetics , Leukemia, Myeloid/genetics , MicroRNAs/genetics , Mutant Chimeric Proteins/genetics , Receptors, Mitogen/genetics , Trans-Splicing , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Differentiation/genetics , Cell Line , Cytarabine/pharmacology , Humans , Lectins, C-Type/metabolism , Leukemia, Myeloid/metabolism , MicroRNAs/metabolism , Mutant Chimeric Proteins/metabolism , Receptors, Mitogen/metabolism , Transcriptional Activation
10.
Metab Eng ; 60: 87-96, 2020 07.
Article in English | MEDLINE | ID: mdl-32268192

ABSTRACT

Miltiradiene is a key intermediate in the biosynthesis of many important natural diterpene compounds with significant pharmacological activity, including triptolide, tanshinones, carnosic acid and carnosol. Sufficient accumulation of miltiradiene is vital for the production of these medicinal compounds. In this study, comprehensive engineering strategies were applied to construct a high-yielding miltiradiene producing yeast strain. First, a chassis strain that can accumulate 2.1 g L-1 geranylgeraniol was constructed. Then, diterpene synthases from various species were evaluated for their ability to produce miltiradiene, and a chimeric miltiradiene synthase, consisting of class II diterpene synthase (di-TPS) CfTPS1 from Coleus forskohlii (Plectranthus barbatus) and class I di-TPS SmKSL1 from Salvia miltiorrhiza showed the highest efficiency in the conversion of GGPP to miltiradiene in yeast. Moreover, the miltiradiene yield was further improved by protein modification, which resulted in a final yield of 550.7 mg L-1 in shake flasks and 3.5 g L-1 in a 5-L bioreactor. This work offers an efficient and green process for the production of the important intermediate miltiradiene, and lays a foundation for further pathway reconstruction and the biotechnological production of valuable natural diterpenes.


Subject(s)
Diterpenes/metabolism , Metabolic Engineering/methods , Mutant Chimeric Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Terpenes/metabolism , Biosynthetic Pathways , CRISPR-Cas Systems , Computer Simulation , Diterpenes/chemistry , Fermentation , Metabolic Networks and Pathways , Mutant Chimeric Proteins/genetics , Mutation , Plasmids
11.
Genes Dev ; 26(6): 615-29, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22426537

ABSTRACT

H2A.Y is an essential, divergent Tetrahymena thermophila histone variant. It has a long nonhistone N terminus that contains leucine-rich repeats (LRR) and an LRR cap domain with similarity to Sds22p, a regulator of yeast protein phosphatase 1 (PP1) activity in the nucleus. In growing cells, H2A.Y is incorporated into micronuclei only during S phase, which occurs immediately after micronuclear mitosis. Depletion of H2A.Y causes prolonged retention of mitosis-associated histone H3-S10 phosphorylation and mitotic abnormalities that mimic S10E mutation. In cells where H2A.Y is depleted, an inducible chimeric gene, in which the H2A.Y N terminus is attached to H2A.X, is shown to regulate micronuclear H3-S10 phosphorylation. H2A.Y can also be specifically coimmunoprecipitated with a Tetrahymena PP1 ortholog (Ppo1p). Taken together, these results argue that the N terminus of H2A.Y functions to regulate H3-S10 dephosphorylation. This striking in vivo case of "cross-talk" between a H2A variant and a specific post-translational modification of another histone demonstrates a novel function for a histone variant.


Subject(s)
Histones/metabolism , Mitosis , Mutant Chimeric Proteins/metabolism , Protein Processing, Post-Translational , Tetrahymena thermophila/cytology , Tetrahymena thermophila/metabolism , Amino Acid Sequence , DNA Replication , Gene Knockout Techniques , Histones/classification , Histones/genetics , Molecular Sequence Data , Mutant Chimeric Proteins/classification , Mutant Chimeric Proteins/genetics , Nucleosomes/metabolism , Phosphorylation , Phylogeny , Protein Phosphatase 1/metabolism , Protein Structure, Tertiary , Saccharomyces cerevisiae Proteins/metabolism , ras Proteins/metabolism
12.
Biochem Biophys Res Commun ; 515(2): 386-393, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31155288

ABSTRACT

Chickens, one of the most important industrial animals, are a biological animal model. Here we focused on the transient receptor potential vanilloid 1 (TRPV1) to understand the pain system for acidic stimuli in chickens compared with mice. By using a whole-cell patch clamp system, we confirmed that acidic stimuli activate both chicken TRPV1 (cTRPV1) and mouse TRPV1 (mTRPV1), but the peak current of cTRPV1 is lower than that of mTRPV1, and it is difficult to desensitize cTRPV1 with an acidic stimulus compared to mTRPV1. Since the C-terminal of the calmodulin (CaM) binding site in TRPV1 was reported as one of the important structures for TRPV1 desensitization, we made chimeric cTRPV1 in which the CaM binding site of chicken is changed to that of mouse (cTRPV1-mCaM). We also compared the acidic responses of native chicken dorsal root ganglion (DRG) cells with that of mouse DRG cells. The TRPV1-mCaM results showed that the desensitization of mutant cTRPV1 was similar to that of mTRPV1, and that the basal activities of mutant cTRPV1 were significantly higher than those of cTRPV1. It was also difficult to desensitize the chicken DRG cells with an acidic stimulus, unlike the mouse DRG cells. These results suggest that there are differences in the pain transduction systems for acidic stimuli between chickens and mice that are caused by the dysfunction of the C-terminal CaM biding site of cTRPV1. These results imply that chickens repeatedly feel weak pain from an acidic stimulus, without desensitization.


Subject(s)
Avian Proteins/metabolism , TRPV Cation Channels/metabolism , Amino Acid Sequence , Animals , Avian Proteins/chemistry , Avian Proteins/genetics , Binding Sites/genetics , Calmodulin/metabolism , Capsaicin/pharmacology , Cells, Cultured , Chickens , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Patch-Clamp Techniques , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Species Specificity , TRPV Cation Channels/chemistry , TRPV Cation Channels/genetics
13.
J Biol Chem ; 292(34): 14026-14038, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28684420

ABSTRACT

Hydrogen sulfide (H2S) is a signaling molecule that is toxic at elevated concentrations. In eukaryotes, it is cleared via a mitochondrial sulfide oxidation pathway, which comprises sulfide quinone oxidoreductase, persulfide dioxygenase (PDO), rhodanese, and sulfite oxidase and converts H2S to thiosulfate and sulfate. Natural fusions between the non-heme iron containing PDO and rhodanese, a thiol sulfurtransferase, exist in some bacteria. However, little is known about the role of the PDO-rhodanese fusion (PRF) proteins in sulfur metabolism. Herein, we report the kinetic properties and the crystal structure of a PRF from the Gram-negative endophytic bacterium Burkholderia phytofirmans The crystal structures of wild-type PRF and a sulfurtransferase-inactivated C314S mutant with and without glutathione were determined at 1.8, 2.4, and 2.7 Å resolution, respectively. We found that the two active sites are distant and do not show evidence of direct communication. The B. phytofirmans PRF exhibited robust PDO activity and preferentially catalyzed sulfur transfer in the direction of thiosulfate to sulfite and glutathione persulfide; sulfur transfer in the reverse direction was detectable only under limited turnover conditions. Together with the kinetic data, our bioinformatics analysis reveals that B. phytofirmans PRF is poised to metabolize thiosulfate to sulfite in a sulfur assimilation pathway rather than in sulfide stress response as seen, for example, with the Staphylococcus aureus PRF or sulfide oxidation and disposal as observed with the homologous mammalian proteins.


Subject(s)
Bacterial Proteins/metabolism , Burkholderiaceae/enzymology , Models, Molecular , Mutant Chimeric Proteins/metabolism , Quinone Reductases/metabolism , Thiosulfate Sulfurtransferase/metabolism , Amino Acid Substitution , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biocatalysis , Catalytic Domain , Computational Biology , Crystallography, X-Ray , Cysteine/chemistry , Disulfides/metabolism , Enzyme Stability , Glutathione/analogs & derivatives , Glutathione/chemistry , Glutathione/metabolism , Hydrogen Sulfide/metabolism , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/genetics , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Quinone Reductases/chemistry , Quinone Reductases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thiosulfate Sulfurtransferase/chemistry , Thiosulfate Sulfurtransferase/genetics , Thiosulfates/metabolism
14.
Mol Ther ; 25(5): 1107-1116, 2017 05 03.
Article in English | MEDLINE | ID: mdl-28392162

ABSTRACT

Viruses can be engineered or adapted for selective propagation in neoplastic tissues and further modified for therapeutic transgene expression to enhance their antitumor potency and druggability. Oncolytic viruses (OVs) can be administered locally or intravenously and spread to a variable degree at sites of tumor growth. OV-infected tumor cells die in situ, releasing viral and tumor antigens that are phagocytosed by macrophages, transported to regional lymph nodes, and presented to antigen-reactive T cells, which proliferate before dispersing to kill uninfected tumor cells at distant sites. Several OVs are showing clinical promise, and one of them, talimogene laherparepvec (T-VEC), was recently granted marketing approval for intratumoral therapy of nonresectable metastatic melanoma. T-VEC also appears to substantially enhance clinical responsiveness to checkpoint inhibitor antibody therapy. Here, we examine the T-VEC paradigm and review some of the approaches currently being pursued to develop the next generation of OVs for both local and systemic administration, as well as for use in combination with other immunomodulatory agents.


Subject(s)
Melanoma/therapy , Multiple Myeloma/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Plasmacytoma/therapy , Skin Neoplasms/therapy , Antigen Presentation , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Humans , Lymph Nodes/immunology , Lymph Nodes/pathology , Measles virus/genetics , Measles virus/metabolism , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Multiple Myeloma/genetics , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Oncolytic Virotherapy/trends , Oncolytic Viruses/immunology , Plasmacytoma/genetics , Plasmacytoma/immunology , Plasmacytoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
15.
Biochem J ; 474(3): 357-376, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27903760

ABSTRACT

In the kidney, the epithelial sodium channel (ENaC) regulates blood pressure through control of sodium and volume homeostasis, and in the lung, ENaC regulates the volume of airway and alveolar fluids. ENaC is a heterotrimer of homologous α-, ß- and γ-subunits, and assembles in the endoplasmic reticulum (ER) before it traffics to and functions at the plasma membrane. Improperly folded or orphaned ENaC subunits are subject to ER quality control and targeted for ER-associated degradation (ERAD). We previously established that a conserved, ER lumenal, molecular chaperone, Lhs1/GRP170, selects αENaC, but not ß- or γ-ENaC, for degradation when the ENaC subunits were individually expressed. We now find that when all three subunits are co-expressed, Lhs1-facilitated ERAD was blocked. To determine which domain-domain interactions between the ENaC subunits are critical for chaperone-dependent quality control, we employed a yeast model and expressed chimeric α/ßENaC constructs in the context of the ENaC heterotrimer. We discovered that the ßENaC transmembrane domain was sufficient to prevent the Lhs1-dependent degradation of the α-subunit in the context of the ENaC heterotrimer. Our work also found that Lhs1 delivers αENaC for proteasome-mediated degradation after the protein has become polyubiquitinated. These data indicate that the Lhs1 chaperone selectively recognizes an immature form of αENaC, one which has failed to correctly assemble with the other channel subunits via its transmembrane domain.


Subject(s)
Endoplasmic Reticulum-Associated Degradation , Epithelial Sodium Channels/metabolism , HSP70 Heat-Shock Proteins/metabolism , Mutant Chimeric Proteins/metabolism , Protein Subunits/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Amino Acid Sequence , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Epithelial Sodium Channels/chemistry , Epithelial Sodium Channels/genetics , Gene Expression , HSP70 Heat-Shock Proteins/chemistry , HSP70 Heat-Shock Proteins/genetics , Humans , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Domains , Protein Folding , Protein Multimerization , Protein Subunits/chemistry , Protein Subunits/genetics , Proteolysis , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Ubiquitination
16.
Biochemistry ; 56(14): 2010-2023, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28362483

ABSTRACT

Terpenoid synthases catalyze isoprenoid cyclization reactions underlying the generation of more than 80,000 natural products. Such dramatic chemodiversity belies the fact that these enzymes generally consist of only three domain folds designated as α, ß, and γ. Catalysis by class I terpenoid synthases occurs exclusively in the α domain, which is found with α, αα, αß, and αßγ domain architectures. Here, we explore the influence of domain architecture on catalysis by taxadiene synthase from Taxus brevifolia (TbTS, αßγ), fusicoccadiene synthase from Phomopsis amygdali (PaFS, (αα)6), and ophiobolin F synthase from Aspergillus clavatus (AcOS, αα). We show that the cyclization fidelity and catalytic efficiency of the α domain of TbTS are severely compromised by deletion of the ßγ domains; however, retention of the ß domain preserves significant cyclization fidelity. In PaFS, we previously demonstrated that one α domain similarly influences catalysis by the other α domain [ Chen , M. , Chou , W. K. W. , Toyomasu , T. , Cane , D. E. , and Christianson , D. W. ( 2016 ) ACS Chem. Biol. 11 , 889 - 899 ]. Here, we show that the hexameric quaternary structure of PaFS enables cluster channeling. We also show that the α domains of PaFS and AcOS can be swapped so as to make functional chimeric αα synthases. Notably, both cyclization fidelity and catalytic efficiency are altered in all chimeric synthases. Twelve newly formed and uncharacterized C20 diterpene products and three C25 sesterterpene products are generated by these chimeras. Thus, engineered αßγ and αα terpenoid cyclases promise to generate chemodiversity in the greater family of terpenoid natural products.


Subject(s)
Alkyl and Aryl Transferases/chemistry , Aspergillus/genetics , Isomerases/chemistry , Mutant Chimeric Proteins/chemistry , Saccharomycetales/genetics , Taxus/genetics , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Aspergillus/enzymology , Cyclization , Diterpenes/metabolism , Gene Expression , Isomerases/genetics , Isomerases/metabolism , Kinetics , Models, Molecular , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Protein Domains , Protein Engineering , Protein Structure, Secondary , Saccharomycetales/enzymology , Sesterterpenes/biosynthesis , Taxus/enzymology
17.
Biochim Biophys Acta Bioenerg ; 1858(11): 927-938, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28826909

ABSTRACT

The reaction centre-light harvesting 1 (RC-LH1) complex of Thermochromatium (Tch.) tepidum has a unique calcium-ion binding site that enhances thermal stability and red-shifts the absorption of LH1 from 880nm to 915nm in the presence of calcium-ions. The LH1 antenna of mesophilic species of phototrophic bacteria such as Rhodobacter (Rba.) sphaeroides does not possess such properties. We have engineered calcium-ion binding into the LH1 antenna of Rba. sphaeroides by progressively modifying the native LH1 polypeptides with sequences from Tch. tepidum. We show that acquisition of the C-terminal domains from LH1 α and ß of Tch. tepidum is sufficient to activate calcium-ion binding and the extent of red-shifting increases with the proportion of Tch. tepidum sequence incorporated. However, full exchange of the LH1 polypeptides with those of Tch. tepidum results in misassembled core complexes. Isolated α and ß polypeptides from our most successful mutant were reconstituted in vitro with BChl a to form an LH1-type complex, which was stabilised 3-fold by calcium-ions. Additionally, carotenoid specificity was changed from spheroidene found in Rba. sphaeroides to spirilloxanthin found in Tch. tepidum, with the latter enhancing in vitro formation of LH1. These data show that the C-terminal LH1 α/ß domains of Tch. tepidum behave autonomously, and are able to transmit calcium-ion induced conformational changes to BChls bound to the rest of a foreign antenna complex. Thus, elements of foreign antenna complexes, such as calcium-ion binding and blue/red switching of absorption, can be ported into Rhodobacter sphaeroides using careful design processes.


Subject(s)
Bacterial Proteins/chemistry , Calcium/chemistry , Chromatiaceae/chemistry , Mutant Chimeric Proteins/chemistry , Photosynthetic Reaction Center Complex Proteins/chemistry , Rhodobacter sphaeroides/genetics , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Calcium/metabolism , Carotenoids/chemistry , Carotenoids/metabolism , Cations, Divalent , Chromatiaceae/metabolism , Gene Expression , Genetic Engineering , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Photosynthetic Reaction Center Complex Proteins/genetics , Photosynthetic Reaction Center Complex Proteins/metabolism , Protein Binding , Rhodobacter sphaeroides/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Xanthophylls/chemistry , Xanthophylls/metabolism
18.
Plant J ; 85(5): 675-85, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26846460

ABSTRACT

Efficient stomatal opening requires activation of KAT-type K(+) channels, which mediate K(+) influx into guard cells. Most KAT-type channels are functionally facilitated by extracellular acidification. However, despite sequence and structural homologies, the maize counterpart of Arabidopsis KAT1 (ZmK2.1) is resistant to pH activation. To understand the structural determinant that results in the differential pH activation of these counterparts, we analysed chimeric channels and channels with point mutations for ZmK2.1 and its closest Arabidopsis homologue KAT1. Exchange of the S1-S2 linkers altered the pH sensitivity between the two channels, suggesting that the S1-S2 linker is essentially involved in the pH sensitivity. The effects of D92 mutation within the linker motif together with substitution of the first half of the linker largely resemble the effects of substitution of the complete linker. Topological modelling predicts that one of the two cysteines located on the outer face section of the S5 domain may serve as a potential titratable group that interacts with the S1-S2 linker. The difference between ZmK2.1 and KAT1 is predicted to be the result of the distance of the stabilized linkers from the titratable group. In KAT1, residue K85 within the linker forms a hydrogen bond with C211 that enables the pH activation; conversely, the linker of ZmK2.1 is distantly located and thus does not interact with the equivalent titration group (C208). Thus, in addition to the known structural contributors to the proton activation of KAT channels, we have uncovered a previously unidentified component that is strongly involved in this complex proton activation network.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/genetics , Plant Proteins/genetics , Potassium Channels, Inwardly Rectifying/genetics , Zea mays/genetics , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/genetics , Amino Acids/metabolism , Arabidopsis/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Binding Sites/genetics , Hydrogen Bonding , Hydrogen-Ion Concentration , Models, Molecular , Mutant Chimeric Proteins/chemistry , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Mutation , Plant Proteins/chemistry , Plant Proteins/metabolism , Potassium Channels, Inwardly Rectifying/chemistry , Potassium Channels, Inwardly Rectifying/metabolism , Protein Binding , Protein Domains , Sequence Homology, Amino Acid , Zea mays/metabolism
19.
Neurobiol Dis ; 99: 24-35, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27988344

ABSTRACT

Axon regeneration in the central nervous system is limited both by inhibitory extracellular cues and by an intrinsically low capacity for axon growth in some CNS populations. Chondroitin sulfate proteoglycans (CSPGs) are well-studied inhibitors of axon growth in the CNS, and degradation of CSPGs by chondroitinase has been shown to improve the extension of injured axons. Alternatively, axon growth can be improved by targeting the neuron-intrinsic growth capacity through forced expression of regeneration-associated transcription factors. For example, a transcriptionally active chimera of Krüppel-like Factor 7 (KLF7) and a VP16 domain improves axon growth when expressed in corticospinal tract neurons. Here we tested the hypothesis that combined expression of chondroitinase and VP16-KLF7 would lead to further improvements in axon growth after spinal injury. Chondroitinase was expressed by viral transduction of cells in the spinal cord, while VP16-KLF7 was virally expressed in sensory neurons of the dorsal root ganglia or corticospinal tract (CST) neurons. After transection of the dorsal columns, both chondroitinase and VP16-KLF7 increased the proximity of severed sensory axons to the injury site. Similarly, after complete crush injuries, VP16-KLF7 expression increased the approach of CST axons to the injury site. In neither paradigm however, did single or combined treatment with chondroitinase or VP16-KLF7 enable regenerative growth distal to the injury. These results substantiate a role for CSPG inhibition and low KLF7 activity in determining the net retraction of axons from sites of spinal injury, while suggesting that additional factors act to limit a full regenerative response.


Subject(s)
Axons/metabolism , Chondroitin ABC Lyase/administration & dosage , Kruppel-Like Transcription Factors/administration & dosage , Neurons, Afferent/metabolism , Pyramidal Tracts/metabolism , Spinal Cord Injuries/therapy , Animals , Axons/pathology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chondroitin ABC Lyase/genetics , Chondroitin ABC Lyase/metabolism , Disease Models, Animal , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Genetic Therapy , Genetic Vectors , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice, Inbred C57BL , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Neuronal Outgrowth/physiology , Neurons, Afferent/pathology , Proteus vulgaris , Pyramidal Tracts/pathology , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
20.
Blood ; 125(22): 3466-76, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25887778

ABSTRACT

T cells expressing a chimeric antigen receptor (CAR) can produce dramatic results in lymphocytic leukemia patients; however, therapeutic strategies for myeloid leukemia remain limited. Folate receptor ß (FRß) is a myeloid-lineage antigen expressed on 70% of acute myeloid leukemia (AML) patient samples. Here, we describe the development and evaluation of the first CARs specific for human FRß (m909) in vitro and in vivo. m909 CAR T cells exhibited selective activation and lytic function against engineered C30-FRß as well as endogenous FRß(+) AML cell lines in vitro. In mouse models of human AML, m909 CAR T cells mediated the regression of engrafted FRß(+) THP1 AML in vivo. In addition, we demonstrated that treatment of AML with all-trans retinoic acid (ATRA) enhanced FRß expression, resulting in improved immune recognition by m909 CAR T cells. Because many cell surface markers are shared between AML blasts and healthy hematopoietic stem and progenitor cells (HSCs), we evaluated FRß expression and recognition of HSCs by CAR T cells. m909 CAR T cells were not toxic against healthy human CD34(+) HSCs in vitro. Our results indicate that FRß is a promising target for CAR T-cell therapy of AML, which may be augmented by combination with ATRA.


Subject(s)
Folate Receptor 2/antagonists & inhibitors , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Animals , Cells, Cultured , Female , Folate Receptor 2/genetics , Genetic Therapy/methods , HEK293 Cells , Humans , Immunotherapy, Adoptive/methods , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Molecular Targeted Therapy , Mutant Chimeric Proteins/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL