31,014 results on '"Protein Subunits"'
Search Results
2. Subtype-specific conformational landscape of NMDA receptor gating.
- Author
-
Bleier, Julia, Furtado de Mendonca, Philipe, Habrian, Chris, Stanley, Cherise, Vyklicky, Vojtech, and Isacoff, Ehud
- Subjects
CP: Neuroscience ,FRET ,GluN1 ,GluN2 ,Grin1 ,Grin2 ,NMDA receptors ,allostery ,single-molecule FRET ,Receptors ,N-Methyl-D-Aspartate ,Humans ,Fluorescence Resonance Energy Transfer ,Animals ,Protein Conformation ,HEK293 Cells ,Ion Channel Gating ,Protein Subunits ,Protein Domains - Abstract
N-methyl-D-aspartate receptors are ionotropic glutamate receptors that mediate synaptic transmission and plasticity. Variable GluN2 subunits in diheterotetrameric receptors with identical GluN1 subunits set very different functional properties. To understand this diversity, we use single-molecule fluorescence resonance energy transfer (smFRET) to measure the conformations of the ligand binding domain and modulatory amino-terminal domain of the common GluN1 subunit in receptors with different GluN2 subunits. Our results demonstrate a strong influence of the GluN2 subunits on GluN1 rearrangements, both in non-agonized and partially agonized activation intermediates, which have been elusive to structural analysis, and in the fully liganded state. Chimeric analysis reveals structural determinants that contribute to these subtype differences. Our study provides a framework for understanding the conformational landscape that supports highly divergent levels of activity, desensitization, and agonist potency in receptors with different GluN2s and could open avenues for the development of subtype-specific modulators.
- Published
- 2024
3. How much does TRPV1 deviate from an ideal MWC-type protein?
- Author
-
Li, Shisheng and Zheng, Jie
- Subjects
TRPV Cation Channels ,Ion Channel Gating ,Models ,Molecular ,Ligands ,Protein Subunits ,Animals ,Protein Binding ,Models ,Biological ,Capsaicin - Abstract
Many ion channels are known to behave as an allosteric protein, coupling environmental stimuli captured by specialized sensing domains to the opening of a central pore. The classic Monod-Wyman-Changeux (MWC) model, originally proposed to describe binding of gas molecules to hemoglobin, has been widely used as a framework for analyzing ion channel gating. Here, we address the issue of how accurately the MWC model predicts activation of the capsaicin receptor TRPV1 by vanilloids. Taking advantage of a concatemeric design that makes it possible to lock TRPV1 in states with zero to four bound vanilloid molecules, we showed quantitatively that the overall gating behavior is satisfactorily predicted by the MWC model. There is, however, a small yet detectable subunit position effect: ligand binding to two kitty-corner subunits is 0.3-0.4 kcal/mol more effective in inducing opening than binding to two neighbor subunits. This difference-less than 10% of the overall energetic contribution from ligand binding-might be due to the restriction on subunit arrangement imposed by the planar membrane; if this is the case, then the position effect is not expected in hemoglobin, in which each subunit is related equivalently to all the other subunits.
- Published
- 2024
4. Precisely patterned nanofibres made from extendable protein multiplexes
- Author
-
Bethel, Neville P, Borst, Andrew J, Parmeggiani, Fabio, Bick, Matthew J, Brunette, TJ, Nguyen, Hannah, Kang, Alex, Bera, Asim K, Carter, Lauren, Miranda, Marcos C, Kibler, Ryan D, Lamb, Mila, Li, Xinting, Sankaran, Banumathi, and Baker, David
- Subjects
Chemical Sciences ,Nanofibers ,Models ,Molecular ,Protein Conformation ,alpha-Helical ,Protein Subunits ,Organic Chemistry ,Chemical sciences - Abstract
Molecular systems with coincident cyclic and superhelical symmetry axes have considerable advantages for materials design as they can be readily lengthened or shortened by changing the length of the constituent monomers. Among proteins, alpha-helical coiled coils have such symmetric, extendable architectures, but are limited by the relatively fixed geometry and flexibility of the helical protomers. Here we describe a systematic approach to generating modular and rigid repeat protein oligomers with coincident C2 to C8 and superhelical symmetry axes that can be readily extended by repeat propagation. From these building blocks, we demonstrate that a wide range of unbounded fibres can be systematically designed by introducing hydrophilic surface patches that force staggering of the monomers; the geometry of such fibres can be precisely tuned by varying the number of repeat units in the monomer and the placement of the hydrophilic patches.
- Published
- 2023
5. Identification of unique α4 chain structure and conserved antiangiogenic activity of α3NC1 type IV collagen in zebrafish.
- Author
-
LeBleu, Valerie, Dai, Jianli, MacDonald, Brian, Alge, Joseph, Sund, Malin, Xie, Liang, Sugimoto, Hikaru, Tainer, John, Zon, Leonard, Kalluri, Raghu, and Tsutakawa, Susan
- Subjects
Tumstatin ,angiogenesis ,basement membrane ,collagen IV ,zebrafish ,Animals ,Humans ,Collagen Type IV ,Zebrafish ,Endothelial Cells ,Protein Subunits ,Basement Membrane - Abstract
BACKGROUND: Type IV collagen is an abundant component of basement membranes in all multicellular species and is essential for the extracellular scaffold supporting tissue architecture and function. Lower organisms typically have two type IV collagen genes, encoding α1 and α2 chains, in contrast with the six genes in humans, encoding α1-α6 chains. The α chains assemble into trimeric protomers, the building blocks of the type IV collagen network. The detailed evolutionary conservation of type IV collagen network remains to be studied. RESULTS: We report on the molecular evolution of type IV collagen genes. The zebrafish α4 non-collagenous (NC1) domain, in contrast with its human ortholog, contains an additional cysteine residue and lacks the M93 and K211 residues involved in sulfilimine bond formation between adjacent protomers. This may alter α4 chain interactions with other α chains, as supported by temporal and anatomic expression patterns of collagen IV chains during the zebrafish development. Despite the divergence between zebrafish and human α3 NC1 domain (endogenous angiogenesis inhibitor, Tumstatin), the zebrafish α3 NC1 domain exhibits conserved antiangiogenic activity in human endothelial cells. CONCLUSIONS: Our work supports type IV collagen is largely conserved between zebrafish and humans, with a possible difference involving the α4 chain.
- Published
- 2023
6. Chemical shift assignments of calmodulin bound to a C-terminal site (residues 1120–1147) in the β-subunit of a retinal cyclic nucleotide-gated channel (CNGB1)
- Author
-
Bej, Aritra and Ames, James B
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neurosciences ,Calcium ,Calmodulin ,Cyclic Nucleotide-Gated Cation Channels ,Humans ,Nuclear Magnetic Resonance ,Biomolecular ,Nucleotides ,Cyclic ,Protein Subunits ,Retinal Rod Photoreceptor Cells ,CaM ,CNGB1 ,Retina ,Photoreceptor ,NMR ,Biophysics ,Biochemistry and cell biology - Abstract
Retinal cyclic nucleotide-gated (CNG) channels consist of two protein subunits (CNGA1 and CNGB1). Calmodulin (CaM) binds to two separate sites within the cytosolic region of CNGB1: CaM binding to an N-terminal site (human CNGB1 residues 565-587, called CaM1) decreases the open probability of CNG channels at elevated Ca2+ levels in dark-adapted photoreceptors, whereas CaM binding to a separate C-terminal site (CNGB1 residues 1120-1147, called CaM2) may increase channel open probability in light activated photoreceptors. We recently reported NMR chemical shift assignments of Ca2+-saturated CaM bound to the CaM1 site of CNGB1 (BMRB no. 51222). Here, we report complete NMR chemical shift assignments of Ca2+-saturated CaM bound to the C-terminal CaM2 site of CNGB1 (BMRB no. 51447).
- Published
- 2022
7. Kv Channel Ancillary Subunits: Where Do We Go from Here?
- Author
-
Abbott, Geoffrey W
- Subjects
Underpinning research ,1.1 Normal biological development and functioning ,Humans ,Potassium ,Potassium Channels ,Potassium Channels ,Voltage-Gated ,Protein Subunits ,cardiac arrhythmia ,KCNE ,KCNQ ,MinK-related peptide ,potassium channel ,Physiology ,Human Movement and Sports Sciences ,Medical Physiology ,Biochemistry & Molecular Biology - Abstract
Voltage-gated potassium (Kv) channels each comprise four pore-forming α-subunits that orchestrate essential duties such as voltage sensing and K+ selectivity and conductance. In vivo, however, Kv channels also incorporate regulatory subunits-some Kv channel specific, others more general modifiers of protein folding, trafficking, and function. Understanding all the above is essential for a complete picture of the role of Kv channels in physiology and disease.
- Published
- 2022
8. De novo design of protein homodimers containing tunable symmetric protein pockets
- Author
-
Hicks, Derrick R, Kennedy, Madison A, Thompson, Kirsten A, DeWitt, Michelle, Coventry, Brian, Kang, Alex, Bera, Asim K, Brunette, TJ, Sankaran, Banumathi, Stoddard, Barry, and Baker, David
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Medicinal and Biomolecular Chemistry ,Chemical Sciences ,Built Environment and Design ,Design ,Generic health relevance ,Ligands ,Models ,Molecular ,Protein Binding ,Protein Subunits ,homodimer ,protein design ,repeat protein ,scaffold ,symmetry - Abstract
Function follows form in biology, and the binding of small molecules requires proteins with pockets that match the shape of the ligand. For design of binding to symmetric ligands, protein homo-oligomers with matching symmetry are advantageous as each protein subunit can make identical interactions with the ligand. Here, we describe a general approach to designing hyperstable C2 symmetric proteins with pockets of diverse size and shape. We first designed repeat proteins that sample a continuum of curvatures but have low helical rise, then docked these into C2 symmetric homodimers to generate an extensive range of C2 symmetric cavities. We used this approach to design thousands of C2 symmetric homodimers, and characterized 101 of them experimentally. Of these, the geometry of 31 were confirmed by small angle X-ray scattering and 2 were shown by crystallographic analyses to be in close agreement with the computational design models. These scaffolds provide a rich set of starting points for binding a wide range of C2 symmetric compounds.
- Published
- 2022
9. Sialoglycan-binding patterns of bacterial AB5 toxin B subunits correlate with host range and toxicity, indicating evolution independent of A subunits.
- Author
-
Khan, Naazneen, Sasmal, Aniruddha, Khedri, Zahra, Secrest, Patrick, Verhagen, Andrea, Srivastava, Saurabh, Varki, Nissi, Chen, Xi, Yu, Hai, Beddoe, Travis, Paton, Adrienne W, Paton, James C, and Varki, Ajit
- Subjects
Animals ,Mammals ,Bacteria ,Yersinia pestis ,Plague ,N-Acetylneuraminic Acid ,Polysaccharides ,Protein Subunits ,Bacterial Toxins ,Evolution ,Molecular ,Phylogeny ,Protein Binding ,Host Specificity ,bacterial ,cytotoxicity ,evolution ,host range ,pathogenesis ,phylogenetic ,sialoglycan microarray ,toxin ,Vector-Borne Diseases ,Biodefense ,Prevention ,Emerging Infectious Diseases ,Infectious Diseases ,Vaccine Related ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Chemical Sciences ,Biological Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology - Abstract
Many pathogenic bacteria secrete AB5 toxins that can be virulence factors. Cytotoxic A subunits are delivered to the cytosol following B subunit binding to specific host cell surface glycans. Some B subunits are not associated with A subunits, for example, YpeB of Yersinia pestis, the etiologic agent of plague. Plague cannot be eradicated because of Y. pestis' adaptability to numerous hosts. We previously showed selective binding of other B5 pentamers to a sialoglycan microarray, with sialic acid (Sia) preferences corresponding to those prominently expressed by various hosts, for example, N-acetylneuraminic acid (Neu5Ac; prominent in humans) or N-glycolylneuraminic acid (Neu5Gc; prominent in ruminant mammals and rodents). Here, we report that A subunit phylogeny evolved independently of B subunits and suggest a future B subunit nomenclature based on bacterial species names. We also found via phylogenetic analysis of B subunits, which bind Sias, that homologous molecules show poor correlation with species phylogeny. These data indicate ongoing lateral gene transfers between species, including mixing of A and B subunits. Consistent with much broader host range of Y. pestis, we show that YpeB recognizes all mammalian Sia types, except for 4-O-acetylated ones. Notably, YpeB alone causes dose-dependent cytotoxicity, which is abolished by a mutation (Y77F) eliminating Sia recognition, suggesting that cell proliferation and death are promoted via lectin-like crosslinking of cell surface sialoglycoconjugates. These findings help explain the host range of Y. pestis and could be important for pathogenesis. Overall, our data indicate ongoing rapid evolution of both host Sias and pathogen toxin-binding properties.
- Published
- 2022
10. 新疆不同地区加工专用花生营养成分比较.
- Author
-
罗舒舒, 张雨, 张甜甜, 代蕾, 高建宇, and 王强
- Abstract
Copyright of Modern Food Science & Technology is the property of Editorial Office of Modern Food Science & Technology and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2024
- Full Text
- View/download PDF
11. The efficacy of γ-aminobutyric acid type A receptor (GABA AR) subtype-selective positive allosteric modulators in blocking tetramethylenedisulfotetramine (TETS)-induced seizure-like behavior in larval zebrafish with minimal sedation.
- Author
-
Mundy, Paige C, Pressly, Brandon, Carty, Dennis R, Yaghoobi, Bianca, Wulff, Heike, and Lein, Pamela J
- Subjects
Animals ,Zebrafish ,Seizures ,Midazolam ,Receptors ,GABA-A ,Protein Subunits ,GABA Modulators ,Hypnotics and Sedatives ,Convulsants ,Behavior ,Animal ,Larva ,Locomotion ,Bridged-Ring Compounds ,Benzodiazepine ,Chemical Threat Agent ,GABA(A)R ,Tetramethylenedisulfotetramine ,Toxicology ,Pharmacology and Pharmaceutical Sciences - Abstract
The chemical threat agent tetramethylenedisulfotetramine (TETS) is a γ-aminobutyric acid type A receptor (GABA AR) antagonist that causes life threatening seizures. Currently, there is no specific antidote for TETS intoxication. TETS-induced seizures are typically treated with benzodiazepines, which function as nonselective positive allosteric modulators (PAMs) of synaptic GABAARs. The major target of TETS was recently identified as the GABAAR α2β3γ2 subtype in electrophysiological studies using recombinantly expressed receptor combinations. Here, we tested whether these in vitro findings translate in vivo by comparing the efficacy of GABAAR subunit-selective PAMs in reducing TETS-induced seizure behavior in larval zebrafish. We tested PAMs targeting α1, α2, α2/3/5, α6, ß2/3, ß1/2/3, and δ subunits and compared their efficacy to the benzodiazepine midazolam (MDZ). The data demonstrate that α2- and α6-selective PAMs (SL-651,498 and SB-205384, respectively) were effective at mitigating TETS-induced seizure-like behavior. Combinations of SB-205384 and MDZ or SL-651,498 and 2-261 (ß2/3-selective) mitigated TETS-induced seizure-like behavior at concentrations that did not elicit sedating effects in a photomotor behavioral assay, whereas MDZ alone caused sedation at the concentration required to stop seizure behavior. Isobologram analyses suggested that SB-205384 and MDZ interacted in an antagonistic fashion, while the effects of SL-651,498 and 2-261 were additive. These results further elucidate the molecular mechanism by which TETS induces seizures and provide mechanistic insight regarding specific countermeasures against this chemical convulsant.
- Published
- 2021
12. Accurate prediction of protein structures and interactions using a three-track neural network
- Author
-
Baek, Minkyung, DiMaio, Frank, Anishchenko, Ivan, Dauparas, Justas, Ovchinnikov, Sergey, Lee, Gyu Rie, Wang, Jue, Cong, Qian, Kinch, Lisa N, Schaeffer, R Dustin, Millán, Claudia, Park, Hahnbeom, Adams, Carson, Glassman, Caleb R, DeGiovanni, Andy, Pereira, Jose H, Rodrigues, Andria V, van Dijk, Alberdina A, Ebrecht, Ana C, Opperman, Diederik J, Sagmeister, Theo, Buhlheller, Christoph, Pavkov-Keller, Tea, Rathinaswamy, Manoj K, Dalwadi, Udit, Yip, Calvin K, Burke, John E, Garcia, K Christopher, Grishin, Nick V, Adams, Paul D, Read, Randy J, and Baker, David
- Subjects
Generic health relevance ,ADAM Proteins ,Amino Acid Sequence ,Computer Simulation ,Cryoelectron Microscopy ,Crystallography ,X-Ray ,Databases ,Protein ,Deep Learning ,Membrane Proteins ,Models ,Molecular ,Multiprotein Complexes ,Neural Networks ,Computer ,Protein Conformation ,Protein Folding ,Protein Subunits ,Proteins ,Receptors ,G-Protein-Coupled ,Sphingosine N-Acyltransferase ,General Science & Technology - Abstract
DeepMind presented notably accurate predictions at the recent 14th Critical Assessment of Structure Prediction (CASP14) conference. We explored network architectures that incorporate related ideas and obtained the best performance with a three-track network in which information at the one-dimensional (1D) sequence level, the 2D distance map level, and the 3D coordinate level is successively transformed and integrated. The three-track network produces structure predictions with accuracies approaching those of DeepMind in CASP14, enables the rapid solution of challenging x-ray crystallography and cryo-electron microscopy structure modeling problems, and provides insights into the functions of proteins of currently unknown structure. The network also enables rapid generation of accurate protein-protein complex models from sequence information alone, short-circuiting traditional approaches that require modeling of individual subunits followed by docking. We make the method available to the scientific community to speed biological research.
- Published
- 2021
13. Noncanonical protein kinase A activation by oligomerization of regulatory subunits as revealed by inherited Carney complex mutations
- Author
-
Jafari, Naeimeh, Del Rio, Jason, Akimoto, Madoka, Byun, Jung Ah, Boulton, Stephen, Moleschi, Kody, Alsayyed, Yousif, Swanson, Pascale, Huang, Jinfeng, Martinez Pomier, Karla, Lee, Chi, Wu, Jian, Taylor, Susan S, and Melacini, Giuseppe
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Rare Diseases ,Digestive Diseases ,Genetics ,2.1 Biological and endogenous factors ,Allosteric Regulation ,Animals ,Binding Sites ,Carney Complex ,Cattle ,Crystallography ,X-Ray ,Cyclic AMP ,Cyclic AMP-Dependent Protein Kinase RIalpha Subunit ,Dysostoses ,Enzyme Activation ,Gene Expression ,Humans ,Intellectual Disability ,Kinetics ,Models ,Molecular ,Mutation ,Osteochondrodysplasias ,Protein Binding ,Protein Conformation ,alpha-Helical ,Protein Conformation ,beta-Strand ,Protein Interaction Domains and Motifs ,Protein Multimerization ,Protein Subunits ,Recombinant Proteins ,Substrate Specificity ,cAMP ,PKA ,Carney ,aggregation ,oligomerization - Abstract
Familial mutations of the protein kinase A (PKA) R1α regulatory subunit lead to a generalized predisposition for a wide range of tumors, from pituitary adenomas to pancreatic and liver cancers, commonly referred to as Carney complex (CNC). CNC mutations are known to cause overactivation of PKA, but the molecular mechanisms underlying such kinase overactivity are not fully understood in the context of the canonical cAMP-dependent activation of PKA. Here, we show that oligomerization-induced sequestration of R1α from the catalytic subunit of PKA (C) is a viable mechanism of PKA activation that can explain the CNC phenotype. Our investigations focus on comparative analyses at the level of structure, unfolding, aggregation, and kinase inhibition profiles of wild-type (wt) PKA R1α, the A211D and G287W CNC mutants, as well as the cognate acrodysostosis type 1 (ACRDYS1) mutations A211T and G287E. The latter exhibit a phenotype opposite to CNC with suboptimal PKA activation compared with wt. Overall, our results show that CNC mutations not only perturb the classical cAMP-dependent allosteric activation pathway of PKA, but also amplify significantly more than the cognate ACRDYS1 mutations nonclassical and previously unappreciated activation pathways, such as oligomerization-induced losses of the PKA R1α inhibitory function.
- Published
- 2021
14. Structure of human telomerase holoenzyme with bound telomeric DNA
- Author
-
Ghanim, George E, Fountain, Adam J, van Roon, Anne-Marie M, Rangan, Ramya, Das, Rhiju, Collins, Kathleen, and Nguyen, Thi Hoang Duong
- Subjects
Genetics ,2.1 Biological and endogenous factors ,Aetiology ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Binding Sites ,Catalytic Domain ,Cryoelectron Microscopy ,DNA ,Histones ,Holoenzymes ,Humans ,Models ,Molecular ,Mutation ,Nucleic Acid Conformation ,Nucleotide Motifs ,Protein Subunits ,RNA ,Ribonucleoproteins ,Telomerase ,Telomere ,General Science & Technology - Abstract
Telomerase adds telomeric repeats at chromosome ends to compensate for the telomere loss that is caused by incomplete genome end replication1. In humans, telomerase is upregulated during embryogenesis and in cancers, and mutations that compromise the function of telomerase result in disease2. A previous structure of human telomerase at a resolution of 8 Å revealed a vertebrate-specific composition and architecture3, comprising a catalytic core that is flexibly tethered to an H and ACA (hereafter, H/ACA) box ribonucleoprotein (RNP) lobe by telomerase RNA. High-resolution structural information is necessary to develop treatments that can effectively modulate telomerase activity as a therapeutic approach against cancers and disease. Here we used cryo-electron microscopy to determine the structure of human telomerase holoenzyme bound to telomeric DNA at sub-4 Å resolution, which reveals crucial DNA- and RNA-binding interfaces in the active site of telomerase as well as the locations of mutations that alter telomerase activity. We identified a histone H2A-H2B dimer within the holoenzyme that was bound to an essential telomerase RNA motif, which suggests a role for histones in the folding and function of telomerase RNA. Furthermore, this structure of a eukaryotic H/ACA RNP reveals the molecular recognition of conserved RNA and protein motifs, as well as interactions that are crucial for understanding the molecular pathology of many mutations that cause disease. Our findings provide the structural details of the assembly and active site of human telomerase, which paves the way for the development of therapeutic agents that target this enzyme.
- Published
- 2021
15. Protomer alignment modulates specificity of RNA substrate recognition by Ire1.
- Author
-
Li, Weihan, Crotty, Kelly, Garrido Ruiz, Diego, Voorhies, Mark, Rivera, Carlos, Sil, Anita, Mullins, R Dyche, Jacobson, Matthew P, Peschek, Jirka, and Walter, Peter
- Subjects
Ire1 ,RNA biology ,S. cerevisiae ,S. pombe ,biochemistry ,chemical biology ,enzymatic substrate specificity ,unfolded protein response ,Membrane Glycoproteins ,Molecular Dynamics Simulation ,Phylogeny ,Protein Subunits ,Protein-Serine-Threonine Kinases ,RNA ,RNA Splicing ,Ribonucleases ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,Schizosaccharomyces ,Sequence Alignment ,Substrate Specificity ,Genetics ,1.1 Normal biological development and functioning ,Biochemistry and Cell Biology - Abstract
The unfolded protein response (UPR) maintains protein folding homeostasis in the endoplasmic reticulum (ER). In metazoan cells, the Ire1 branch of the UPR initiates two functional outputs-non-conventional mRNA splicing and selective mRNA decay (RIDD). By contrast, Ire1 orthologs from Saccharomyces cerevisiae and Schizosaccharomyces pombe are specialized for only splicing or RIDD, respectively. Previously, we showed that the functional specialization lies in Ire1's RNase activity, which is either stringently splice-site specific or promiscuous (Li et al., 2018). Here, we developed an assay that reports on Ire1's RNase promiscuity. We found that conversion of two amino acids within the RNase domain of S. cerevisiae Ire1 to their S. pombe counterparts rendered it promiscuous. Using biochemical assays and computational modeling, we show that the mutations rewired a pair of salt bridges at Ire1 RNase domain's dimer interface, changing its protomer alignment. Thus, Ire1 protomer alignment affects its substrates specificity.
- Published
- 2021
16. Structure of the human Mediator-bound transcription preinitiation complex
- Author
-
Abdella, R, Talyzina, A, Chen, S, Inouye, CJ, Tjian, R, and He, Y
- Subjects
Genetics ,Generic health relevance ,Binding Sites ,Catalytic Domain ,Cryoelectron Microscopy ,Cyclin-Dependent Kinases ,Humans ,Mediator Complex ,Models ,Molecular ,Phosphorylation ,Protein Binding ,Protein Domains ,Protein Subunits ,RNA Polymerase II ,Transcription Factor TFIIH ,Transcription Factors ,General ,Transcription Initiation ,Genetic ,Cyclin-Dependent Kinase-Activating Kinase ,General Science & Technology - Abstract
Eukaryotic transcription requires the assembly of a multisubunit preinitiation complex (PIC) composed of RNA polymerase II (Pol II) and the general transcription factors. The coactivator Mediator is recruited by transcription factors, facilitates the assembly of the PIC, and stimulates phosphorylation of the Pol II C-terminal domain (CTD) by the TFIIH subunit CDK7. Here, we present the cryo-electron microscopy structure of the human Mediator-bound PIC at a resolution below 4 angstroms. Transcription factor binding sites within Mediator are primarily flexibly tethered to the tail module. CDK7 is stabilized by multiple contacts with Mediator. Two binding sites exist for the Pol II CTD, one between the head and middle modules of Mediator and the other in the active site of CDK7, providing structural evidence for Pol II CTD phosphorylation within the Mediator-bound PIC.
- Published
- 2021
17. Stiffness of Nanoparticulate Mineralized Collagen Scaffolds Triggers Osteogenesis via Mechanotransduction and Canonical Wnt Signaling
- Author
-
Zhou, Qi, Lyu, Shengyu, Bertrand, Anthony A, Hu, Allison C, Chan, Candace H, Ren, Xiaoyan, Dewey, Marley J, Tiffany, Aleczandria S, Harley, Brendan AC, and Lee, Justine C
- Subjects
Engineering ,Biomedical Engineering ,Stem Cell Research ,Regenerative Medicine ,Stem Cell Research - Nonembryonic - Human ,Dental/Oral and Craniofacial Disease ,Actins ,Adaptor Proteins ,Signal Transducing ,Bone Morphogenetic Protein 2 ,Bone Morphogenetic Protein Receptors ,Cell Nucleus ,Collagen ,Core Binding Factor Alpha 1 Subunit ,Cross-Linking Reagents ,Cytosol ,Focal Adhesion Protein-Tyrosine Kinases ,Gene Expression Regulation ,Glycosaminoglycans ,Humans ,Integrins ,Intracellular Signaling Peptides and Proteins ,Mechanotransduction ,Cellular ,Mesenchymal Stem Cells ,Minerals ,Models ,Biological ,Nanoparticles ,Osteogenesis ,Phosphorylation ,Polymerization ,Protein Subunits ,Smad Proteins ,Tissue Scaffolds ,Transcription Factors ,Transcriptional Coactivator with PDZ-Binding Motif Proteins ,Wnt Signaling Pathway ,YAP-Signaling Proteins ,beta Catenin ,rho GTP-Binding Proteins ,β ,‐ ,catenin ,mechanotransduction ,scaffolds ,Wnt ,YAP ,TAZ ,YAP/TAZ ,β-catenin ,Macromolecular and Materials Chemistry ,Chemical Engineering ,Polymers ,Macromolecular and materials chemistry ,Biomedical engineering - Abstract
The ability of the extracellular matrix (ECM) to instruct progenitor cell differentiation has generated excitement for the development of materials-based regenerative solutions. Described a nanoparticulate mineralized collagen glycosaminoglycan (MC-GAG) material capable of inducing in vivo skull regeneration without exogenous growth factors or ex vivo progenitor cell-priming is described previously. Here, the contribution of titrating stiffness to osteogenicity is evaluated by comparing noncrosslinked (NX-MC) and crosslinked (MC) forms of MC-GAG. While both materials are osteogenic, MC demonstrates an increased expression of osteogenic markers and mineralization compared to NX-MC. Both materials are capable of autogenously activating the canonical BMPR signaling pathway with phosphorylation of Smad1/5. However, unlike NX-MC, human mesenchymal stem cells cultured on MC demonstrate significant elevations in the major mechanotransduction mediators YAP and TAZ expression, coincident with β-catenin activation in the canonical Wnt signaling pathway. Inhibition of YAP/TAZ activation reduces osteogenic expression, mineralization, and β-catenin activation in MC, with less of an effect on NX-MC. YAP/TAZ inhibition also results in a reciprocal increase in Smad1/5 phosphorylation and BMP2 expression. The results indicate that increasing MC-GAG stiffness induces osteogenic differentiation via the mechanotransduction mediators YAP/TAZ and the canonical Wnt signaling pathway, whereas the canonical BMPR signaling pathway is activated independent of stiffness.
- Published
- 2021
18. Involvement of the Microglial Aryl Hydrocarbon Receptor in Neuroinflammation and Vasogenic Edema after Ischemic Stroke
- Author
-
Tanaka, Miki, Fujikawa, Masaho, Oguro, Ami, Itoh, Kouichi, Vogel, Christoph FA, and Ishihara, Yasuhiro
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Brain Disorders ,Stroke ,Cerebrovascular ,2.1 Biological and endogenous factors ,Animals ,Brain Edema ,Brain Injuries ,Cytochrome P-450 CYP1A1 ,Cytokines ,Encephalitis ,Humans ,Indoleamine-Pyrrole 2 ,3 ,-Dioxygenase ,Infarction ,Middle Cerebral Artery ,Ischemic Stroke ,Ligands ,Macrophages ,Male ,Mice ,Inbred ICR ,Microglia ,NADPH Oxidases ,Oxidative Stress ,Promoter Regions ,Genetic ,Protein Subunits ,Reactive Oxygen Species ,Receptors ,Aryl Hydrocarbon ,THP-1 Cells ,Tumor Necrosis Factor-alpha ,Up-Regulation ,Mice ,ischemia ,edema ,AhR ,inflammation ,p47phox ,Biological sciences ,Biomedical and clinical sciences - Abstract
Microglia are activated after ischemic stroke and induce neuroinflammation. The expression of the aryl hydrocarbon receptor (AhR) has recently been reported to elicit cytokine expression. We previously reported that microglial activation mediates ischemic edema progression. Thus, the purpose of this study was to examine the role of AhR in inflammation and edema after ischemia using a mouse middle cerebral artery occlusion (MCAO) model. MCAO upregulated AhR expression in microglia during ischemia. MCAO increased the expression of tumor necrosis factor α (TNFα) and then induced edema progression, and worsened the modified neurological severity scores, with these being suppressed by administration of an AhR antagonist, CH223191. In THP-1 macrophages, the NADPH oxidase (NOX) subunit p47phox was significantly increased by AhR ligands, especially under inflammatory conditions. Suppression of NOX activity by apocynin or elimination of superoxide by superoxide dismutase decreased TNFα expression, which was induced by the AhR ligand. AhR ligands also elicited p47phox expression in mouse primary microglia. Thus, p47phox may be important in oxidative stress and subsequent inflammation. In MCAO model mice, P47phox expression was upregulated in microglia by ischemia. Lipid peroxidation induced by MCAO was suppressed by CH223191. Taken together, these findings suggest that AhR in the microglia is involved in neuroinflammation and subsequent edema, after MCAO via p47phox expression upregulation and oxidative stress.
- Published
- 2021
19. U2 snRNA structure is influenced by SF3A and SF3B proteins but not by SF3B inhibitors
- Author
-
Urabe, Veronica K, Stevers, Meredith, Ghosh, Arun K, and Jurica, Melissa S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Generic health relevance ,HeLa Cells ,Humans ,Nucleic Acid Conformation ,Protein Structure ,Secondary ,Protein Subunits ,RNA ,Small Nuclear ,Ribonucleoprotein ,U2 Small Nuclear ,Spliceosomes ,Hela Cells ,General Science & Technology - Abstract
U2 snRNP is an essential component of the spliceosome. It is responsible for branch point recognition in the spliceosome A-complex via base-pairing of U2 snRNA with an intron to form the branch helix. Small molecule inhibitors target the SF3B component of the U2 snRNP and interfere with A-complex formation during spliceosome assembly. We previously found that the first SF3B inhibited-complex is less stable than A-complex and hypothesized that SF3B inhibitors interfere with U2 snRNA secondary structure changes required to form the branch helix. Using RNA chemical modifiers, we probed U2 snRNA structure in A-complex and SF3B inhibited splicing complexes. The reactivity pattern for U2 snRNA in the SF3B inhibited-complex is indistinguishable from that of A-complex, suggesting that they have the same secondary structure conformation, including the branch helix. This observation suggests SF3B inhibited-complex instability does not stem from an alternate RNA conformation and instead points to the inhibitors interfering with protein component interactions that normally stabilize U2 snRNP's association with an intron. In addition, we probed U2 snRNA in the free U2 snRNP in the presence of SF3B inhibitor and again saw no differences. However, increased protection of nucleotides upstream of Stem I in the absence of SF3A and SF3B proteins suggests a change of secondary structure at the very 5' end of U2 snRNA. Chemical probing of synthetic U2 snRNA in the absence of proteins results in similar protections and predicts a previously uncharacterized extension of Stem I. Because this stem must be disrupted for SF3A and SF3B proteins to stably join the snRNP, the structure has the potential to influence snRNP assembly and recycling after spliceosome disassembly.
- Published
- 2021
20. Functional NMDA receptors are expressed by human pulmonary artery smooth muscle cells
- Author
-
Dong, Yi Na, Hsu, Fu-Chun, Koziol-White, Cynthia J, Stepanova, Victoria, Jude, Joseph, Gritsiuta, Andrei, Rue, Ryan, Mott, Rosalind, Coulter, Douglas A, Panettieri, Reynold A, Krymskaya, Vera P, Takano, Hajime, Goncharova, Elena A, Goncharov, Dmitry A, Cines, Douglas B, and Lynch, David R
- Subjects
Neurosciences ,Genetics ,Lung ,Cardiovascular ,Animals ,Cells ,Cultured ,Humans ,Mice ,Mice ,Inbred C57BL ,Muscle ,Smooth ,Vascular ,Myocytes ,Smooth Muscle ,Protein Subunits ,Pulmonary Artery ,Receptors ,N-Methyl-D-Aspartate ,Vasoconstriction - Abstract
N-methyl-D-aspartate (NMDA) receptors are widely expressed in the central nervous system. However, their presence and function at extraneuronal sites is less well characterized. In the present study, we examined the expression of NMDA receptor subunit mRNA and protein in human pulmonary artery (HPA) by quantitative polymerase chain reaction (PCR), immunohistochemistry and immunoblotting. We demonstrate that both GluN1 and GluN2 subunit mRNAs are expressed in HPA. In addition, GluN1 and GluN2 (A-D) subunit proteins are expressed by human pulmonary artery smooth muscle cells (HPASMCs) in vitro and in vivo. These subunits localize on the surface of HPASMCs and form functional ion channels as evidenced by whole-cell patch-clamp electrophysiology and reduced phenylephrine-induced contractile responsiveness of human pulmonary artery by the NMDA receptor antagonist MK801 under hypoxic condition. HPASMCs also express high levels of serine racemase and vesicular glutamate transporter 1, suggesting a potential source of endogenous agonists for NMDA receptor activation. Our findings show HPASMCs express functional NMDA receptors in line with their effect on pulmonary vasoconstriction, and thereby suggest a novel therapeutic target for pharmacological modulations in settings associated with pulmonary vascular dysfunction.
- Published
- 2021
21. Stoichiometry of Nucleotide Binding to Proteasome AAA+ ATPase Hexamer Established by Native Mass Spectrometry
- Author
-
Yu, Yadong, Liu, Haichuan, Yu, Zanlin, Witkowska, H Ewa, and Cheng, Yifan
- Subjects
ATPases Associated with Diverse Cellular Activities ,Adenosine Diphosphate ,Adenylyl Imidodiphosphate ,Archaeal Proteins ,Ligands ,Mass Spectrometry ,Methanocaldococcus ,Mutant Proteins ,Nucleotides ,Proteasome Endopeptidase Complex ,Protein Binding ,Protein Multimerization ,Protein Subunits ,Spectrometry ,Mass ,Electrospray Ionization ,proteasome ,AAA plus ATPase ,nucleotide binding ,stoichiometry ,native mass spectrometry ,cooperativity ,mass spectrometry ,Archaebacteria* ,electron microscopy ,macromolecular complex analysis ,non-covalent interaction MS* ,AAA+ ATPase ,Biochemistry & Molecular Biology - Abstract
AAA+ ATPases constitute a large family of proteins that are involved in a plethora of cellular processes including DNA disassembly, protein degradation and protein complex disassembly. They typically form a hexametric ring-shaped structure with six subunits in a (pseudo) 6-fold symmetry. In a subset of AAA+ ATPases that facilitate protein unfolding and degradation, six subunits cooperate to translocate protein substrates through a central pore in the ring. The number and type of nucleotides in an AAA+ ATPase hexamer is inherently linked to the mechanism that underlies cooperation among subunits and couples ATP hydrolysis with substrate translocation. We conducted a native MS study of a monodispersed form of PAN, an archaeal proteasome AAA+ ATPase, to determine the number of nucleotides bound to each hexamer of the WT protein. We utilized ADP and its analogs (TNP-ADP and mant-ADP), and a nonhydrolyzable ATP analog (AMP-PNP) to study nucleotide site occupancy within the PAN hexamer in ADP- and ATP-binding states, respectively. Throughout all experiments we used a Walker A mutant (PANK217A) that is impaired in nucleotide binding as an internal standard to mitigate the effects of residual solvation on mass measurement accuracy and to serve as a reference protein to control for nonspecific nucleotide binding. This approach led to the unambiguous finding that a WT PAN hexamer carried - from expression host - six tightly bound ADP molecules that could be exchanged for ADP and ATP analogs. Although the Walker A mutant did not bind ADP analogs, it did bind AMP-PNP, albeit at multiple stoichiometries. We observed variable levels of hexamer dissociation and an appearance of multimeric species with the over-charged molecular ion distributions across repeated experiments. We posit that these phenomena originated during ESI process at the final stages of ESI droplet evolution.
- Published
- 2020
22. Sarcolipin Exhibits Abundant RNA Transcription and Minimal Protein Expression in Horse Gluteal Muscle.
- Author
-
Autry, Joseph M, Karim, Christine B, Perumbakkam, Sudeep, Finno, Carrie J, McKenzie, Erica C, Thomas, David D, and Valberg, Stephanie J
- Subjects
equidae ,gene expression profiling ,intracellular membranes ,long noncoding RNA ,peptides ,protein subunits ,rhabdomyolysis ,sarcolipin ,sarcoplasmic reticulum calcium-transporting ATPases ,western blotting ,Veterinary Sciences - Abstract
Ca2+ regulation in equine muscle is important for horse performance, yet little is known about this species-specific regulation. We reported recently that horse encode unique gene and protein sequences for the sarcoplasmic reticulum (SR) Ca2+-transporting ATPase (SERCA) and the regulatory subunit sarcolipin (SLN). Here we quantified gene transcription and protein expression of SERCA and its inhibitory peptides in horse gluteus, as compared to commonly-studied rabbit skeletal muscle. RNA sequencing and protein immunoblotting determined that horse gluteus expresses the ATP2A1 gene (SERCA1) as the predominant SR Ca2+-ATPase isoform and the SLN gene as the most-abundant SERCA inhibitory peptide, as also found in rabbit skeletal muscle. Equine muscle expresses an insignificant level of phospholamban (PLN), another key SERCA inhibitory peptide expressed commonly in a variety of mammalian striated muscles. Surprisingly in horse, the RNA transcript ratio of SLN-to-ATP2A1 is an order of magnitude higher than in rabbit, while the corresponding protein expression ratio is an order of magnitude lower than in rabbit. Thus, SLN is not efficiently translated or maintained as a stable protein in horse muscle, suggesting a non-coding role for supra-abundant SLN mRNA. We propose that the lack of SLN and PLN inhibition of SERCA activity in equine muscle is an evolutionary adaptation that potentiates Ca2+ cycling and muscle contractility in a prey species domestically selected for speed.
- Published
- 2020
23. Scalable continuous evolution for the generation of diverse enzyme variants encompassing promiscuous activities.
- Author
-
Rix, Gordon, Watkins-Dulaney, Ella J, Almhjell, Patrick J, Boville, Christina E, Arnold, Frances H, and Liu, Chang C
- Subjects
Thermotoga maritima ,Tryptophan Synthase ,Tryptophan ,Bacterial Proteins ,Protein Subunits ,Evolution ,Molecular ,Substrate Specificity ,Mutation ,Biocatalysis ,Evolution ,Molecular - Abstract
Enzyme orthologs sharing identical primary functions can have different promiscuous activities. While it is possible to mine this natural diversity to obtain useful biocatalysts, generating comparably rich ortholog diversity is difficult, as it is the product of deep evolutionary processes occurring in a multitude of separate species and populations. Here, we take a first step in recapitulating the depth and scale of natural ortholog evolution on laboratory timescales. Using a continuous directed evolution platform called OrthoRep, we rapidly evolve the Thermotoga maritima tryptophan synthase β-subunit (TmTrpB) through multi-mutation pathways in many independent replicates, selecting only on TmTrpB's primary activity of synthesizing L-tryptophan from indole and L-serine. We find that the resulting sequence-diverse TmTrpB variants span a range of substrate profiles useful in industrial biocatalysis and suggest that the depth and scale of evolution that OrthoRep affords will be generally valuable in enzyme engineering and the evolution of biomolecular functions.
- Published
- 2020
24. Structures of capsid and capsid-associated tegument complex inside the Epstein–Barr virus
- Author
-
Liu, Wei, Cui, Yanxiang, Wang, Caiyan, Li, Zihang, Gong, Danyang, Dai, Xinghong, Bi, Guo-Qiang, Sun, Ren, and Zhou, Z Hong
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,Lymphatic Research ,Cancer ,Hematology ,Lymphoma ,Rare Diseases ,2.2 Factors relating to the physical environment ,2.1 Biological and endogenous factors ,Infection ,Amino Acid Sequence ,Capsid ,Capsid Proteins ,Cryoelectron Microscopy ,Epstein-Barr Virus Infections ,Herpesvirus 4 ,Human ,Humans ,Imaging ,Three-Dimensional ,Models ,Molecular ,Protein Subunits ,Structure-Activity Relationship ,Virion ,Virus Assembly ,Microbiology ,Medical Microbiology - Abstract
As the first discovered human cancer virus, Epstein-Barr virus (EBV) causes Burkitt's lymphoma and nasopharyngeal carcinoma. Isolating virions for determining high-resolution structures has been hindered by latency-a hallmark of EBV infection-and atomic structures are thus available only for recombinantly expressed EBV proteins. In the present study, by symmetry relaxation and subparticle reconstruction, we have determined near-atomic-resolution structures of the EBV capsid with an asymmetrically attached DNA-translocating portal and capsid-associated tegument complexes from cryogenic electron microscopy images of just 2,048 EBV virions obtained by chemical induction. The resulting atomic models reveal structural plasticity among the 20 conformers of the major capsid protein, 2 conformers of the small capsid protein (SCP), 4 conformers of the triplex monomer proteins and 2 conformers of the triplex dimer proteins. Plasticity reaches the greatest level at the capsid-tegument interfaces involving SCP and capsid-associated tegument complexes (CATC): SCPs crown pentons/hexons and mediate tegument protein binding, and CATCs bind and rotate all five periportal triplexes, but notably only about one peri-penton triplex. These results offer insights into the EBV capsid assembly and a mechanism for recruiting cell-regulating factors into the tegument compartment as 'cargoes', and should inform future anti-EBV strategies.
- Published
- 2020
25. Structure of human GABAB receptor in an inactive state
- Author
-
Park, Jinseo, Fu, Ziao, Frangaj, Aurel, Liu, Jonathan, Mosyak, Lidia, Shen, Tong, Slavkovich, Vesna N, Ray, Kimberly M, Taura, Jaume, Cao, Baohua, Geng, Yong, Zuo, Hao, Kou, Yongjun, Grassucci, Robert, Chen, Shaoxia, Liu, Zheng, Lin, Xin, Williams, Justin P, Rice, William J, Eng, Edward T, Huang, Rick K, Soni, Rajesh K, Kloss, Brian, Yu, Zhiheng, Javitch, Jonathan A, Hendrickson, Wayne A, Slesinger, Paul A, Quick, Matthias, Graziano, Joseph, Yu, Hongtao, Fiehn, Oliver, Clarke, Oliver B, Frank, Joachim, and Fan, Qing R
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neurosciences ,1.1 Normal biological development and functioning ,Neurological ,Generic health relevance ,Calcium ,Cryoelectron Microscopy ,Ethanolamines ,Humans ,Ligands ,Models ,Molecular ,Phosphorylcholine ,Protein Domains ,Protein Multimerization ,Protein Subunits ,Receptors ,GABA-B ,Structure-Activity Relationship ,General Science & Technology - Abstract
The human GABAB receptor-a member of the class C family of G-protein-coupled receptors (GPCRs)-mediates inhibitory neurotransmission and has been implicated in epilepsy, pain and addiction1. A unique GPCR that is known to require heterodimerization for function2-6, the GABAB receptor has two subunits, GABAB1 and GABAB2, that are structurally homologous but perform distinct and complementary functions. GABAB1 recognizes orthosteric ligands7,8, while GABAB2 couples with G proteins9-14. Each subunit is characterized by an extracellular Venus flytrap (VFT) module, a descending peptide linker, a seven-helix transmembrane domain and a cytoplasmic tail15. Although the VFT heterodimer structure has been resolved16, the structure of the full-length receptor and its transmembrane signalling mechanism remain unknown. Here we present a near full-length structure of the GABAB receptor, captured in an inactive state by cryo-electron microscopy. Our structure reveals several ligands that preassociate with the receptor, including two large endogenous phospholipids that are embedded within the transmembrane domains to maintain receptor integrity and modulate receptor function. We also identify a previously unknown heterodimer interface between transmembrane helices 3 and 5 of both subunits, which serves as a signature of the inactive conformation. A unique 'intersubunit latch' within this transmembrane interface maintains the inactive state, and its disruption leads to constitutive receptor activity.
- Published
- 2020
26. Effect of magnitude and variability of energy of activation in multisite ultrasensitive biochemical processes.
- Author
-
Lagunes, Leonila, Bardwell, Lee, and Enciso, German A
- Subjects
Proteins ,Protein Subunits ,Ligands ,Signal Transduction ,Protein Processing ,Post-Translational ,Binding Sites ,Protein Conformation ,Energy Metabolism ,Phosphorylation ,Thermodynamics ,Models ,Biological ,Models ,Biological ,Protein Processing ,Post-Translational ,Bioinformatics ,Mathematical Sciences ,Biological Sciences ,Information and Computing Sciences - Abstract
Protein activity is often regulated by ligand binding or by post-translational modifications such as phosphorylation. Moreover, proteins that are regulated in this way often contain multiple ligand binding sites or modification sites, which can operate to create an ultrasensitive dose response. Here, we consider the contribution of the individual modification/binding sites to the activation process, and how their individual values affect the ultrasensitive behavior of the overall system. We use a generalized Monod-Wyman-Changeux (MWC) model that allows for variable conformational free energy contributions from distinct sites, and associate a so-called activation parameter to each site. Our analysis shows that the ultrasensitivity generally increases as the conformational free energy contribution from one or more sites is strengthened. Furthermore, ultrasensitivity depends on the mean of the activation parameters and not on their variability. In some cases, we find that the best way to maximize ultrasensitivity is to make the contribution from all sites as strong as possible. These results provide insights into the performance objectives of multiple modification/binding sites and thus help gain a greater understanding of signaling and its role in diseases.
- Published
- 2020
27. A Family of Auxiliary Subunits of the TRP Cation Channel Encoded by the Complex inaF Locus.
- Author
-
Chen, Zijing and Montell, Craig
- Subjects
Drosophila melanogaster ,TRP channel ,inaF ,photoreceptor cells ,phototransduction ,β subunit ,Adaptor Proteins ,Signal Transducing ,Animals ,Drosophila Proteins ,Drosophila melanogaster ,Eye Proteins ,Membrane Potentials ,Photoreceptor Cells ,Invertebrate ,Protein Binding ,Protein Stability ,Protein Subunits ,Protein Transport ,Transient Receptor Potential Channels - Abstract
TRP channels function in many types of sensory receptor cells. Despite extensive analyses, an open question is whether there exists a family of auxiliary subunits, which could influence localization, trafficking, and function of TRP channels. Here, using Drosophila melanogaster, we reveal a previously unknown TRP interacting protein, INAF-C, which is expressed exclusively in the ultraviolet-sensing R7 photoreceptor cells. INAF-C is encoded by an unusual locus comprised of four distinct coding regions, which give rise to four unique single-transmembrane-containing proteins. With the exception of INAF-B, roles for the other INAF proteins were unknown. We found that both INAF-B and INAF-C are required for TRP stability and localization in R7 cells. Conversely, loss of just INAF-B greatly reduced TRP from other types of photoreceptor cells, but not R7. The requirements for TRP and INAF are reciprocal, since loss of TRP decreased the concentrations of both INAF-B and INAF-C. INAF-A, which is not normally expressed in photoreceptor cells, can functionally substitute for INAF-B, indicating that it is a third TRP auxiliary protein. Reminiscent of the structural requirements between Kv channels and KCNE auxiliary subunits, the codependencies of TRP and INAF depended on several transmembrane domains (TMDs) in TRP, and the TMD and the C-terminus of INAF-B. Our studies support a model in which the inaF locus encodes a family of at least three TRP auxiliary subunits.
- Published
- 2020
28. Backbone assignments and conformational dynamics in the S. typhimurium tryptophan synthase α-subunit from solution-state NMR.
- Author
-
Sakhrani, Varun V, Hilario, Eduardo, Caulkins, Bethany G, Hatcher-Skeers, Mary E, Fan, Li, Dunn, Michael F, and Mueller, Leonard J
- Subjects
Salmonella typhimurium ,Nitrogen Isotopes ,Tryptophan Synthase ,Bacterial Proteins ,Protein Subunits ,Solutions ,Crystallography ,X-Ray ,Nuclear Magnetic Resonance ,Biomolecular ,Binding Sites ,Protein Conformation ,Protein Structure ,Secondary ,Catalysis ,Models ,Molecular ,Molecular Dynamics Simulation ,Chemical shift assignments ,Ligand titration ,Protein NMR ,Protein dynamics ,Relaxation ,Tryptophan synthase ,Vaccine Related ,Emerging Infectious Diseases ,Infectious Diseases ,Physical Sciences ,Chemical Sciences ,Biological Sciences ,Biophysics - Abstract
Backbone assignments for the isolated α-subunit of Salmonella typhimurium tryptophan synthase (TS) are reported based on triple resonance solution-state NMR experiments on a uniformly 2H,13C,15N-labeled sample. From the backbone chemical shifts, secondary structure and random coil index order parameters (RCI-S2) are predicted. Titration with the 3-indole-D-glycerol 3'-phosphate analog, N-(4'-trifluoromethoxybenzenesulfonyl)-2-aminoethyl phosphate (F9), leads to chemical shift perturbations indicative of conformational changes from which an estimate of the dissociation constant is obtained. Comparisons of the backbone chemical-shifts, RCI-S2 values, and site-specific relaxation times with and without F9 reveal allosteric changes including modulation in secondary structures and loop rigidity induced upon ligand binding. A comparison is made to the X-ray crystal structure of the α-subunit in the full TS αββα bi-enzyme complex and to two new X-ray crystal structures of the isolated TS α-subunit reported in this work.
- Published
- 2020
29. Ribonucleotide Reductases: Structure, Chemistry, and Metabolism Suggest New Therapeutic Targets
- Author
-
Greene, Brandon L, Kang, Gyunghoon, Cui, Chang, Bennati, Marina, Nocera, Daniel G, Drennan, Catherine L, and Stubbe, JoAnne
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Anti-Bacterial Agents ,Antineoplastic Agents ,Biocatalysis ,Drug Discovery ,Enzyme Inhibitors ,Escherichia coli ,Escherichia coli Infections ,Humans ,Molecular Docking Simulation ,Neoplasms ,Nucleotides ,Oxidation-Reduction ,Protein Structure ,Secondary ,Protein Subunits ,Ribonucleotide Reductases ,Small Molecule Libraries ,Structure-Activity Relationship ,ribonucleotide reductases ,structures ,mechanisms ,therapeutics ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biochemistry and cell biology - Abstract
Ribonucleotide reductases (RNRs) catalyze the de novo conversion of nucleotides to deoxynucleotides in all organisms, controlling their relative ratios and abundance. In doing so, they play an important role in fidelity of DNA replication and repair. RNRs' central role in nucleic acid metabolism has resulted in five therapeutics that inhibit human RNRs. In this review, we discuss the structural, dynamic, and mechanistic aspects of RNR activity and regulation, primarily for the human and Escherichia coli class Ia enzymes. The unusual radical-based organic chemistry of nucleotide reduction, the inorganic chemistry of the essential metallo-cofactor biosynthesis/maintenance, the transport of a radical over a long distance, and the dynamics of subunit interactions all present distinct entry points toward RNR inhibition that are relevant for drug discovery. We describe the current mechanistic understanding of small molecules that target different elements of RNR function, including downstream pathways that lead to cell cytotoxicity. We conclude by summarizing novel and emergent RNR targeting motifs for cancer and antibiotic therapeutics.
- Published
- 2020
30. Integrative structure and function of the yeast exocyst complex
- Author
-
Ganesan, Sai J, Feyder, Michael J, Chemmama, Ilan E, Fang, Fei, Rout, Michael P, Chait, Brian T, Shi, Yi, Munson, Mary, and Sali, Andrej
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Bioengineering ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Cryoelectron Microscopy ,Crystallography ,X-Ray ,Models ,Molecular ,Protein Conformation ,Protein Subunits ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,chemical cross-linking mass spectrometry ,EM ,exocytosis ,integrative modeling ,membrane fusion ,protein cross-linking ,SNAREs ,structural models ,yeast exocyst complex ,Computation Theory and Mathematics ,Other Information and Computing Sciences ,Biophysics ,Biochemistry and cell biology ,Medicinal and biomolecular chemistry - Abstract
Exocyst is an evolutionarily conserved hetero-octameric tethering complex that plays a variety of roles in membrane trafficking, including exocytosis, endocytosis, autophagy, cell polarization, cytokinesis, pathogen invasion, and metastasis. Exocyst serves as a platform for interactions between the Rab, Rho, and Ral small GTPases, SNARE proteins, and Sec1/Munc18 regulators that coordinate spatial and temporal fidelity of membrane fusion. However, its mechanism is poorly described at the molecular level. Here, we determine the molecular architecture of the yeast exocyst complex by an integrative approach, based on a 3D density map from negative-stain electron microscopy (EM) at ~16 Å resolution, 434 disuccinimidyl suberate and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride cross-links from chemical-crosslinking mass spectrometry, and partial atomic models of the eight subunits. The integrative structure is validated by a previously determined cryo-EM structure, cross-links, and distances from in vivo fluorescence microscopy. Our subunit configuration is consistent with the cryo-EM structure, except for Sec5. While not observed in the cryo-EM map, the integrative model localizes the N-terminal half of Sec3 near the Sec6 subunit. Limited proteolysis experiments suggest that the conformation of Exo70 is dynamic, which may have functional implications for SNARE and membrane interactions. This study illustrates how integrative modeling based on varied low-resolution structural data can inform biologically relevant hypotheses, even in the absence of high-resolution data.
- Published
- 2020
31. A long lost key opens an ancient lock: Drosophila Myb causes a synthetic multivulval phenotype in nematodes
- Author
-
Vorster, Paul J, Goetsch, Paul, Wijeratne, Tilini U, Guiley, Keelan Z, Andrejka, Laura, Tripathi, Sarvind, Larson, Braden J, Rubin, Seth M, Strome, Susan, and Lipsick, Joseph S
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Biotechnology ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Amino Acid Sequence ,Animals ,Biological Evolution ,Cell Cycle Proteins ,Drosophila ,Drosophila Proteins ,Evolution ,Molecular ,Gene Expression Regulation ,Genetic Association Studies ,Humans ,Models ,Molecular ,Phenotype ,Phylogeny ,Protein Conformation ,Protein Subunits ,Proto-Oncogene Proteins c-myb ,Structure-Activity Relationship ,Myb ,Development ,Evolution ,Oncogene ,synMuv ,Tumor suppressor ,Other Biological Sciences ,Biological sciences ,Biomedical and clinical sciences ,Environmental sciences - Abstract
The five-protein MuvB core complex is highly conserved in animals. This nuclear complex interacts with RB-family tumor suppressor proteins and E2F-DP transcription factors to form DREAM complexes that repress genes that regulate cell cycle progression and cell fate. The MuvB core complex also interacts with Myb family oncoproteins to form the Myb-MuvB complexes that activate many of the same genes. We show that animal-type Myb genes are present in Bilateria, Cnidaria and Placozoa, the latter including the simplest known animal species. However, bilaterian nematode worms lost their animal-type Myb genes hundreds of millions of years ago. Nevertheless, amino acids in the LIN9 and LIN52 proteins that directly interact with the MuvB-binding domains of human B-Myb and Drosophila Myb are conserved in Caenorhabditiselegans Here, we show that, despite greater than 500 million years since their last common ancestor, the Drosophila melanogaster Myb protein can bind to the nematode LIN9-LIN52 proteins in vitro and can cause a synthetic multivulval (synMuv) phenotype in vivo This phenotype is similar to that caused by loss-of-function mutations in C. elegans synMuvB-class genes including those that encode homologs of the MuvB core, RB, E2F and DP. Furthermore, amino acid substitutions in the MuvB-binding domain of Drosophila Myb that disrupt its functions in vitro and in vivo also disrupt these activities in C. elegans We speculate that nematodes and other animals may contain another protein that can bind to LIN9 and LIN52 in order to activate transcription of genes repressed by DREAM complexes.
- Published
- 2020
32. Action of a minimal contractile bactericidal nanomachine
- Author
-
Ge, Peng, Scholl, Dean, Prokhorov, Nikolai S, Avaylon, Jaycob, Shneider, Mikhail M, Browning, Christopher, Buth, Sergey A, Plattner, Michel, Chakraborty, Urmi, Ding, Ke, Leiman, Petr G, Miller, Jeff F, and Zhou, Z Hong
- Subjects
Biological Sciences ,Chemical Sciences ,Physical Chemistry ,Bioengineering ,Genetics ,Bacteriophage T4 ,Cryoelectron Microscopy ,Crystallography ,X-Ray ,Genes ,Bacterial ,Models ,Molecular ,Protein Subunits ,Pseudomonas aeruginosa ,Pyocins ,Substrate Specificity ,Type VI Secretion Systems ,General Science & Technology - Abstract
R-type bacteriocins are minimal contractile nanomachines that hold promise as precision antibiotics1-4. Each bactericidal complex uses a collar to bridge a hollow tube with a contractile sheath loaded in a metastable state by a baseplate scaffold1,2. Fine-tuning of such nucleic acid-free protein machines for precision medicine calls for an atomic description of the entire complex and contraction mechanism, which is not available from baseplate structures of the (DNA-containing) T4 bacteriophage5. Here we report the atomic model of the complete R2 pyocin in its pre-contraction and post-contraction states, each containing 384 subunits of 11 unique atomic models of 10 gene products. Comparison of these structures suggests the following sequence of events during pyocin contraction: tail fibres trigger lateral dissociation of baseplate triplexes; the dissociation then initiates a cascade of events leading to sheath contraction; and this contraction converts chemical energy into mechanical force to drive the iron-tipped tube across the bacterial cell surface, killing the bacterium.
- Published
- 2020
33. Behaviors of individual microtubules and microtubule populations relative to critical concentrations: dynamic instability occurs when critical concentrations are driven apart by nucleotide hydrolysis
- Author
-
Jonasson, Erin M, Mauro, Ava J, Li, Chunlei, Labuz, Ellen C, Mahserejian, Shant M, Scripture, Jared P, Gregoretti, Ivan V, Alber, Mark, and Goodson, Holly V
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Computer Simulation ,Hydrolysis ,Kinetics ,Microtubules ,Models ,Biological ,Nucleotides ,Polymers ,Protein Subunits ,Tubulin ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
The concept of critical concentration (CC) is central to understanding the behavior of microtubules (MTs) and other cytoskeletal polymers. Traditionally, these polymers are understood to have one CC, measured in multiple ways and assumed to be the subunit concentration necessary for polymer assembly. However, this framework does not incorporate dynamic instability (DI), and there is work indicating that MTs have two CCs. We use our previously established simulations to confirm that MTs have (at least) two experimentally relevant CCs and to clarify the behavior of individuals and populations relative to the CCs. At free subunit concentrations above the lower CC (CCElongation), growth phases of individual filaments can occur transiently; above the higher CC (CCNetAssembly), the population's polymer mass will increase persistently. Our results demonstrate that most experimental CC measurements correspond to CCNetAssembly, meaning that "typical" DI occurs below the concentration traditionally considered necessary for polymer assembly. We report that [free tubulin] at steady state does not equal CCNetAssembly, but instead approaches CCNetAssembly asymptotically as [total tubulin] increases, and depends on the number of stable MT nucleation sites. We show that the degree of separation between CCElongation and CCNetAssembly depends on the rate of nucleotide hydrolysis. This clarified framework helps explain and unify many experimental observations.
- Published
- 2020
34. Functional interactions between posttranslationally modified amino acids of methyl-coenzyme M reductase in Methanosarcina acetivorans.
- Author
-
Nayak, Dipti, Liu, Andi, Agrawal, Neha, Rodriguez-Carerro, Roy, Dong, Shi-Hui, Mitchell, Douglas, Nair, Satish, and Metcalf, William
- Subjects
Amino Acids ,Archaeal Proteins ,Catalytic Domain ,Methanosarcina ,Methylation ,Methyltransferases ,Models ,Molecular ,Mutation ,Operon ,Oxidoreductases ,Phenotype ,Protein Processing ,Post-Translational ,Protein Subunits ,Temperature - Abstract
The enzyme methyl-coenzyme M reductase (MCR) plays an important role in mediating global levels of methane by catalyzing a reversible reaction that leads to the production or consumption of this potent greenhouse gas in methanogenic and methanotrophic archaea. In methanogenic archaea, the alpha subunit of MCR (McrA) typically contains four to six posttranslationally modified amino acids near the active site. Recent studies have identified enzymes performing two of these modifications (thioglycine and 5-[S]-methylarginine), yet little is known about the formation and function of the remaining posttranslationally modified residues. Here, we provide in vivo evidence that a dedicated S-adenosylmethionine-dependent methyltransferase encoded by a gene we designated methylcysteine modification (mcmA) is responsible for formation of S-methylcysteine in Methanosarcina acetivorans McrA. Phenotypic analysis of mutants incapable of cysteine methylation suggests that the S-methylcysteine residue might play a role in adaption to mesophilic conditions. To examine the interactions between the S-methylcysteine residue and the previously characterized thioglycine, 5-(S)-methylarginine modifications, we generated M. acetivorans mutants lacking the three known modification genes in all possible combinations. Phenotypic analyses revealed complex, physiologically relevant interactions between the modified residues, which alter the thermal stability of MCR in a combinatorial fashion that is not readily predictable from the phenotypes of single mutants. High-resolution crystal structures of inactive MCR lacking the modified amino acids were indistinguishable from the fully modified enzyme, suggesting that interactions between the posttranslationally modified residues do not exert a major influence on the static structure of the enzyme but rather serve to fine-tune the activity and efficiency of MCR.
- Published
- 2020
35. PPP2R5D-Related Intellectual Disability and Neurodevelopmental Delay: A Review of the Current Understanding of the Genetics and Biochemical Basis of the Disorder
- Author
-
Biswas, Dayita, Cary, Whitney, and Nolta, Jan A
- Subjects
Biochemistry and Cell Biology ,Genetics ,Biological Sciences ,Mental Health ,Neurodegenerative ,Neurosciences ,Pediatric ,Intellectual and Developmental Disabilities (IDD) ,Autism ,Stem Cell Research ,Brain Disorders ,Aetiology ,2.1 Biological and endogenous factors ,Mental health ,Neurological ,Animals ,Cell Cycle Checkpoints ,Developmental Disabilities ,Disease Models ,Animal ,Humans ,Intellectual Disability ,Neurons ,Polymorphism ,Single Nucleotide ,Protein Phosphatase 2 ,Protein Subunits ,PP2A ,PPP2R5D ,autism spectrum disorders ,intellectual disability ,neurodevelopmental disabilities ,phosphatase ,review ,seizures ,Other Chemical Sciences ,Other Biological Sciences ,Chemical Physics ,Biochemistry and cell biology ,Microbiology ,Medicinal and biomolecular chemistry - Abstract
Protein Phosphatase 2 Regulatory Subunit B' Delta (PPP2R5D)-related intellectual disability (ID) and neurodevelopmental delay results from germline de novo mutations in the PPP2R5D gene. This gene encodes the protein PPP2R5D (also known as the B56 delta subunit), which is an isoform of the subunit family B56 of the enzyme serine/threonine-protein phosphatase 2A (PP2A). Clinical signs include intellectual disability (ID); autism spectrum disorder (ASD); epilepsy; speech problems; behavioral challenges; and ophthalmologic, skeletal, endocrine, cardiac, and genital malformations. The association of defective PP2A activity in the brain with a wide range of severity of ID, along with its role in ASD, Alzheimer's disease, and Parkinson's-like symptoms, have recently generated the impetus for further research into mutations within this gene. PP2A, together with protein phosphatase 1 (PP1), accounts for more than 90% of all phospho-serine/threonine dephosphorylations in different tissues. The specificity for a wide variety of substrates is determined through nearly 100 different PP2A holoenzymes that are formed by at least 23 types of regulatory B subunits, and two isoforms each of the catalytic subunit C and the structural subunit A. In the mammalian brain, PP2A-mediated protein dephosphorylation plays an important role in learning and memory. The PPP2R5D subunit is highly expressed in the brain and the PPP2A-PPP2R5D holoenzyme plays an important role in maintaining neurons and regulating neuronal signaling. From 2015 to 2017, 25 individuals with PPP2R5D-related developmental disorder were diagnosed. Since then, Whole-Exome Sequencing (WES) has helped to identify more unrelated individuals clinically diagnosed with a neurodevelopmental disorder with pathological variants of PPP2R5D. In this review, we discuss the current understanding of the clinical and genetic aspects of the disorder in the context of the known functions of the PP2A-PPP2R5D holoenzyme in the brain, as well as the pathogenic mutations in PPP2R5D that lead to deficient PP2A-PPP2R5D dephosphorylation and their implications during development and in the etiology of autism, Parkinson's disease, Alzheimer's disease, and so forth. In the future, tools such as transgenic animals carrying pathogenic PPP2R5D mutations, and patient-derived induced pluripotent stem cell lines need to be developed in order to fully understand the effects of these mutations on different neural cell types.
- Published
- 2020
36. Reversible phosphorylation of Rpn1 regulates 26S proteasome assembly and function
- Author
-
Liu, Xiaoyan, Xiao, Weidi, Zhang, Yanan, Wiley, Sandra E, Zuo, Tao, Zheng, Yingying, Chen, Natalie, Chen, Lu, Wang, Xiaorong, Zheng, Yawen, Huang, Lan, Lin, Shixian, Murphy, Anne N, Dixon, Jack E, Xu, Ping, and Guo, Xing
- Subjects
Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,CRISPR-Cas Systems ,Cell Line ,Enzyme Assays ,Gene Knock-In Techniques ,Humans ,Membrane Proteins ,Mice ,Mice ,Knockout ,Mice ,Transgenic ,Mitochondria ,Nuclear Proteins ,Oxidative Stress ,Phosphoprotein Phosphatases ,Phosphorylation ,Proteasome Endopeptidase Complex ,Protein Serine-Threonine Kinases ,Protein Subunits ,RNA ,Small Interfering ,Serine ,Trans-Activators ,proteasome ,phosphorylation ,UBLCP1 ,PIM ,genetic code expansion - Abstract
The fundamental importance of the 26S proteasome in health and disease suggests that its function must be finely controlled, and yet our knowledge about proteasome regulation remains limited. Posttranslational modifications, especially phosphorylation, of proteasome subunits have been shown to impact proteasome function through different mechanisms, although the vast majority of proteasome phosphorylation events have not been studied. Here, we have characterized 1 of the most frequently detected proteasome phosphosites, namely Ser361 of Rpn1, a base subunit of the 19S regulatory particle. Using a variety of approaches including CRISPR/Cas9-mediated gene editing and quantitative mass spectrometry, we found that loss of Rpn1-S361 phosphorylation reduces proteasome activity, impairs cell proliferation, and causes oxidative stress as well as mitochondrial dysfunction. A screen of the human kinome identified several kinases including PIM1/2/3 that catalyze S361 phosphorylation, while its level is reversibly controlled by the proteasome-resident phosphatase, UBLCP1. Mechanistically, Rpn1-S361 phosphorylation is required for proper assembly of the 26S proteasome, and we have utilized a genetic code expansion system to directly demonstrate that S361-phosphorylated Rpn1 more readily forms a precursor complex with Rpt2, 1 of the first steps of 19S base assembly. These findings have revealed a prevalent and biologically important mechanism governing proteasome formation and function.
- Published
- 2020
37. Structural analyses of the PKA RIIβ holoenzyme containing the oncogenic DnaJB1-PKAc fusion protein reveal protomer asymmetry and fusion-induced allosteric perturbations in fibrolamellar hepatocellular carcinoma
- Author
-
Lu, Tsan-Wen, Aoto, Phillip C, Weng, Jui-Hung, Nielsen, Cole, Cash, Jennifer N, Hall, James, Zhang, Ping, Simon, Sanford M, Cianfrocco, Michael A, and Taylor, Susan S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Cancer ,Liver Cancer ,Rare Diseases ,Digestive Diseases ,Liver Disease ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Adenosine Triphosphate ,Allosteric Regulation ,Carcinoma ,Hepatocellular ,Cryoelectron Microscopy ,Cyclic AMP ,Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit ,Cyclic AMP-Dependent Protein Kinase RIalpha Subunit ,Cyclic AMP-Dependent Protein Kinases ,HSP40 Heat-Shock Proteins ,Holoenzymes ,Humans ,Liver Neoplasms ,Molecular Dynamics Simulation ,Protein Binding ,Protein Subunits ,Recombinant Fusion Proteins ,Scattering ,Small Angle ,X-Ray Diffraction ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Developmental Biology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
When the J-domain of the heat shock protein DnaJB1 is fused to the catalytic (C) subunit of cAMP-dependent protein kinase (PKA), replacing exon 1, this fusion protein, J-C subunit (J-C), becomes the driver of fibrolamellar hepatocellular carcinoma (FL-HCC). Here, we use cryo-electron microscopy (cryo-EM) to characterize J-C bound to RIIβ, the major PKA regulatory (R) subunit in liver, thus reporting the first cryo-EM structure of any PKA holoenzyme. We report several differences in both structure and dynamics that could not be captured by the conventional crystallography approaches used to obtain prior structures. Most striking is the asymmetry caused by the absence of the second cyclic nucleotide binding (CNB) domain and the J-domain in one of the RIIβ:J-C protomers. Using molecular dynamics (MD) simulations, we discovered that this asymmetry is already present in the wild-type (WT) RIIβ2C2 but had been masked in the previous crystal structure. This asymmetry may link to the intrinsic allosteric regulation of all PKA holoenzymes and could also explain why most disease mutations in PKA regulatory subunits are dominant negative. The cryo-EM structure, combined with small-angle X-ray scattering (SAXS), also allowed us to predict the general position of the Dimerization/Docking (D/D) domain, which is essential for localization and interacting with membrane-anchored A-Kinase-Anchoring Proteins (AKAPs). This position provides a multivalent mechanism for interaction of the RIIβ holoenzyme with membranes and would be perturbed in the oncogenic fusion protein. The J-domain also alters several biochemical properties of the RIIβ holoenzyme: It is easier to activate with cAMP, and the cooperativity is reduced. These results provide new insights into how the finely tuned allosteric PKA signaling network is disrupted by the oncogenic J-C subunit, ultimately leading to the development of FL-HCC.
- Published
- 2020
38. PCR Mutagenesis, Cloning, Expression, Fast Protein Purification Protocols and Crystallization of the Wild Type and Mutant Forms of Tryptophan Synthase.
- Author
-
Hilario, Eduardo, Fan, Li, Mueller, Leonard J, and Dunn, Michael F
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,Emerging Infectious Diseases ,Digestive Diseases ,Catalysis ,Cloning ,Molecular ,Crystallization ,Crystallography ,X-Ray ,Escherichia coli ,Mutagenesis ,Site-Directed ,Mutant Proteins ,Polymerase Chain Reaction ,Protein Subunits ,Recombinant Proteins ,Reproducibility of Results ,Salmonella typhimurium ,Small Ubiquitin-Related Modifier Proteins ,Static Electricity ,Tryptophan Synthase ,Psychology ,Cognitive Sciences ,Biochemistry and cell biology - Abstract
Structural studies with tryptophan synthase (TS) bienzyme complex (α2β2 TS) from Salmonella typhimurium have been performed to better understand its catalytic mechanism, allosteric behavior, and details of the enzymatic transformation of substrate to product in PLP-dependent enzymes. In this work, a novel expression system to produce the isolated α- and isolated β-subunit allowed the purification of high amounts of pure subunits and α2β2 StTS complex from the isolated subunits within 2 days. Purification was carried out by affinity chromatography followed by cleavage of the affinity tag, ammonium sulfate precipitation, and size exclusion chromatography (SEC). To better understand the role of key residues at the enzyme β-site, site-direct mutagenesis was performed in prior structural studies. Another protocol was created to purify the wild type and mutant α2β2 StTS complexes. A simple, fast and efficient protocol using ammonium sulfate fractionation and SEC allowed purification of α2β2 StTS complex in a single day. Both purification protocols described in this work have considerable advantages when compared with previous protocols to purify the same complex using PEG 8000 and spermine to crystalize the α2β2 StTS complex along the purification protocol. Crystallization of wild type and some mutant forms occurs under slightly different conditions, impairing the purification of some mutants using PEG 8000 and spermine. To prepare crystals suitable for x-ray crystallographic studies several efforts were made to optimize crystallization, crystal quality and cryoprotection. The methods presented here should be generally applicable for purification of tryptophan synthase subunits and wild type and mutant α2β2 StTS complexes.
- Published
- 2020
39. Commentary on the recent FSH collection: known knowns and known unknowns
- Author
-
Coss, Djurdjica
- Subjects
Infertility ,Contraception/Reproduction ,1.1 Normal biological development and functioning ,Underpinning research ,Reproductive health and childbirth ,Animals ,Female ,Follicle Stimulating Hormone ,Gene Expression Regulation ,Granulosa Cells ,Humans ,Male ,Ovarian Follicle ,Protein Binding ,Protein Processing ,Post-Translational ,Protein Subunits ,Receptors ,FSH ,Review Literature as Topic ,Sertoli Cells ,Signal Transduction ,Spermatogenesis ,FSH ,pituitary ,gonadotrope ,granulosa cells ,ovary ,Sertoli cells ,glycosylation ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Endocrinology & Metabolism - Abstract
Follicle-stimulating hormone (FSH) is a dimeric glycoprotein secreted by the anterior pituitary gonadotrope that is necessary for reproductive function in mammals. FSH primarily regulates granulosa cells and follicular growth in females, and Sertoli cell function in males. Since its identification in the 1930s and sequencing in the 1970s, significant progress has been made in elucidating its regulation and downstream function. Recent advances provide deeper insight into FSH synthesis, and effects in the gonads suggest potential roles in extragonadal tissues and examine pharmacological approaches and clinical applications in infertility treatment that now affect 18% of couples. These advances were discussed in detail in a number of reviews published in the last 2 years in Endocrinology. In this brief commentary, we summarize these reviews and point to the outstanding questions that should be answered in the near future to bridge a gap in our understanding of this hormone.
- Published
- 2020
40. Structure and dynamics of the ASB9 CUL-RING E3 Ligase
- Author
-
Lumpkin, Ryan J, Baker, Richard W, Leschziner, Andres E, and Komives, Elizabeth A
- Subjects
Biochemistry and Cell Biology ,Chemical Sciences ,Biological Sciences ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Allosteric Regulation ,Creatine Kinase ,Cryoelectron Microscopy ,Cullin Proteins ,Elongin ,Humans ,Models ,Molecular ,Protein Binding ,Protein Subunits ,Structure-Activity Relationship ,Substrate Specificity ,Suppressor of Cytokine Signaling Proteins ,Ubiquitin-Protein Ligases - Abstract
The Cullin 5 (CUL5) Ring E3 ligase uses adaptors Elongins B and C (ELOB/C) to bind different SOCS-box-containing substrate receptors, determining the substrate specificity of the ligase. The 18-member ankyrin and SOCS box (ASB) family is the largest substrate receptor family. Here we report cryo-EM data for the substrate, creatine kinase (CKB) bound to ASB9-ELOB/C, and for full-length CUL5 bound to the RING protein, RBX2, which binds various E2s. To date, no full structures are available either for a substrate-bound ASB nor for CUL5. Hydrogen-deuterium exchange (HDX-MS) mapped onto a full structural model of the ligase revealed long-range allostery extending from the substrate through CUL5. We propose a revised allosteric mechanism for how CUL-E3 ligases function. ASB9 and CUL5 behave as rigid rods, connected through a hinge provided by ELOB/C transmitting long-range allosteric crosstalk from the substrate through CUL5 to the RBX2 flexible linker.
- Published
- 2020
41. Cryo-EM structure of the mitochondrial protein-import channel TOM complex at near-atomic resolution
- Author
-
Tucker, Kyle and Park, Eunyong
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Generic health relevance ,Carrier Proteins ,Cryoelectron Microscopy ,Mitochondria ,Mitochondrial Membrane Transport Proteins ,Mitochondrial Precursor Protein Import Complex Proteins ,Models ,Molecular ,Protein Conformation ,Protein Multimerization ,Protein Subunits ,Protein Transport ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,Chemical Sciences ,Medical and Health Sciences ,Biophysics ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Nearly all mitochondrial proteins are encoded by the nuclear genome and imported into mitochondria after synthesis on cytosolic ribosomes. These precursor proteins are translocated into mitochondria by the TOM complex, a protein-conducting channel in the mitochondrial outer membrane. We have determined high-resolution cryo-EM structures of the core TOM complex from Saccharomyces cerevisiae in dimeric and tetrameric forms. Dimeric TOM consists of two copies each of five proteins arranged in two-fold symmetry: pore-forming β-barrel protein Tom40 and four auxiliary α-helical transmembrane proteins. The pore of each Tom40 has an overall negatively charged inner surface attributed to multiple functionally important acidic patches. The tetrameric complex is essentially a dimer of dimeric TOM, which may be capable of forming higher-order oligomers. Our study reveals the detailed molecular organization of the TOM complex and provides new insights about the mechanism of protein translocation into mitochondria.
- Published
- 2019
42. Maturation of the respiratory complex II flavoprotein
- Author
-
Sharma, Pankaj, Maklashina, Elena, Cecchini, Gary, and Iverson, TM
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Underpinning research ,Generic health relevance ,Animals ,Cell Respiration ,Electron Transport Chain Complex Proteins ,Electron Transport Complex II ,Flavin-Adenine Dinucleotide ,Flavoproteins ,Humans ,Protein Binding ,Protein Subunits ,Structure-Activity Relationship ,Medicinal and Biomolecular Chemistry ,Biophysics ,Biochemistry and cell biology - Abstract
Respiratory complexes are complicated multi-subunit cofactor-containing machines that allow cells to harvest energy from the environment. Maturation of these complexes requires protein folding, cofactor insertion, and assembly of multiple subunits into a final, functional complex. Because the intermediate states in complex maturation are transitory, these processes are poorly understood. This review gives an overview of the process of maturation in respiratory complex II with a focus on recent structural studies on intermediates formed during covalent flavinylation of the catalytic subunit, SDHA. Covalent flavinylation has an evolutionary significance because variants of complex II enzymes with the covalent ligand removed by mutagenesis cannot oxidize succinate, but can still perform the reverse reaction and reduce fumarate. Since succinate oxidation is a key step of aerobic respiration, the covalent bond of complex II appears to be important for aerobic life.
- Published
- 2019
43. Phosphorylation of the HCN channel auxiliary subunit TRIP8b is altered in an animal model of temporal lobe epilepsy and modulates channel function
- Author
-
Foote, Kendall M, Lyman, Kyle A, Han, Ye, Michailidis, Ioannis E, Heuermann, Robert J, Mandikian, Danielle, Trimmer, James S, Swanson, Geoffrey T, and Chetkovich, Dane M
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Neurosciences ,Brain Disorders ,Neurodegenerative ,Epilepsy ,2.1 Biological and endogenous factors ,Aetiology ,Neurological ,Amino Acid Sequence ,Animals ,Brain ,Calcium-Calmodulin-Dependent Protein Kinase Type 2 ,Dendrites ,Disease Models ,Animal ,Epilepsy ,Temporal Lobe ,Female ,HEK293 Cells ,Humans ,Ion Channel Gating ,Kainic Acid ,Membrane Proteins ,Mice ,Inbred C57BL ,Peroxins ,Phosphorylation ,Phosphoserine ,Protein Subunits ,Rats ,Sprague-Dawley ,Reproducibility of Results ,ion channel ,epilepsy ,phosphorylation ,protein?protein interaction ,Ca2+ ,calmodulin-dependent protein kinase II ,channelopathy ,HCN ,neuronal excitability ,TLE ,TRIP8b ,Ca2+/calmodulin-dependent protein kinase II ,protein–protein interaction ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Temporal lobe epilepsy (TLE) is a prevalent neurological disorder with many patients experiencing poor seizure control with existing anti-epileptic drugs. Thus, novel insights into the mechanisms of epileptogenesis and identification of new drug targets can be transformative. Changes in ion channel function have been shown to play a role in generating the aberrant neuronal activity observed in TLE. Previous work demonstrates that hyperpolarization-activated cyclic nucleotide-gated (HCN) channels regulate neuronal excitability and are mislocalized within CA1 pyramidal cells in a rodent model of TLE. The subcellular distribution of HCN channels is regulated by an auxiliary subunit, tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b), and disruption of this interaction correlates with channel mislocalization. However, the molecular mechanisms responsible for HCN channel dysregulation in TLE are unclear. Here we investigated whether changes in TRIP8b phosphorylation are sufficient to alter HCN channel function. We identified a phosphorylation site at residue Ser237 of TRIP8b that enhances binding to HCN channels and influences channel gating by altering the affinity of TRIP8b for the HCN cytoplasmic domain. Using a phosphospecific antibody, we demonstrate that TRIP8b phosphorylated at Ser237 is enriched in CA1 distal dendrites and that phosphorylation is reduced in the kainic acid model of TLE. Overall, our findings indicate that the TRIP8b-HCN interaction can be modulated by changes in phosphorylation and suggest that loss of TRIP8b phosphorylation may affect HCN channel properties during epileptogenesis. These results highlight the potential of drugs targeting posttranslational modifications to restore TRIP8b phosphorylation to reduce excitability in TLE.
- Published
- 2019
44. Dynamic BAF chromatin remodeling complex subunit inclusion promotes temporally distinct gene expression programs in cardiogenesis
- Author
-
Hota, Swetansu K, Johnson, Jeffrey R, Verschueren, Erik, Thomas, Reuben, Blotnick, Aaron M, Zhu, Yiwen, Sun, Xin, Pennacchio, Len A, Krogan, Nevan J, and Bruneau, Benoit G
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Genetics ,Heart Disease ,Cardiovascular ,1.1 Normal biological development and functioning ,Animals ,Cell Differentiation ,Chromatin ,Chromatin Assembly and Disassembly ,DNA Helicases ,Gene Expression Regulation ,Developmental ,Genome ,Heart ,Mice ,Multiprotein Complexes ,Myocytes ,Cardiac ,Nuclear Proteins ,Organogenesis ,Protein Binding ,Protein Subunits ,Time Factors ,Transcription Factors ,Differentiation ,Gene regulation ,Medical and Health Sciences ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
Chromatin remodeling complexes instruct cellular differentiation and lineage specific transcription. The BRG1/BRM-associated factor (BAF) complexes are important for several aspects of differentiation. We show that the catalytic subunit gene Brg1 has a specific role in cardiac precursors (CPs) to initiate cardiac gene expression programs and repress non-cardiac expression. Using immunopurification with mass spectrometry, we have determined the dynamic composition of BAF complexes during mammalian cardiac differentiation, identifying several cell-type specific subunits. We focused on the CP- and cardiomyocyte (CM)-enriched subunits BAF60c (SMARCD3) and BAF170 (SMARCC2). Baf60c and Baf170 co-regulate gene expression with Brg1 in CPs, and in CMs their loss results in broadly deregulated cardiac gene expression. BRG1, BAF60c and BAF170 modulate chromatin accessibility, to promote accessibility at activated genes while closing chromatin at repressed genes. BAF60c and BAF170 are required for proper BAF complex composition, and BAF170 loss leads to retention of BRG1 at CP-specific sites. Thus, dynamic interdependent BAF complex subunit assembly modulates chromatin states and thereby participates in directing temporal gene expression programs in cardiogenesis.
- Published
- 2019
45. Two PKA RIα holoenzyme states define ATP as an isoform-specific orthosteric inhibitor that competes with the allosteric activator, cAMP
- Author
-
Lu, Tsan-Wen, Wu, Jian, Aoto, Phillip C, Weng, Jui-Hung, Ahuja, Lalima G, Sun, Nicholas, Cheng, Cecilia Y, Zhang, Ping, and Taylor, Susan S
- Subjects
Biochemistry and Cell Biology ,Medical Physiology ,Biomedical and Clinical Sciences ,Biological Sciences ,1.1 Normal biological development and functioning ,Adenosine Triphosphate ,Allosteric Regulation ,Amino Acid Sequence ,Crystallography ,X-Ray ,Cyclic AMP ,Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit ,Cyclic AMP-Dependent Protein Kinase RIalpha Subunit ,Cyclic AMP-Dependent Protein Kinases ,Gene Expression Regulation ,Enzymologic ,Holoenzymes ,Humans ,Protein Binding ,Protein Structure ,Quaternary ,Protein Subunits ,Signal Transduction ,protein kinase A ,structural biology ,allosteric and orthosteric regulation ,isoform-specific quaternary structure ,cAMP - Abstract
Protein kinase A (PKA) holoenzyme, comprised of a cAMP-binding regulatory (R)-subunit dimer and 2 catalytic (C)-subunits, is the master switch for cAMP-mediated signaling. Of the 4 R-subunits (RIα, RIβ, RIIα, RIIβ), RIα is most essential for regulating PKA activity in cells. Our 2 RIα2C2 holoenzyme states, which show different conformations with and without ATP, reveal how ATP/Mg2+ functions as a negative orthosteric modulator. Biochemical studies demonstrate how the removal of ATP primes the holoenzyme for cAMP-mediated activation. The opposing competition between ATP/cAMP is unique to RIα. In RIIβ, ATP serves as a substrate and facilitates cAMP-activation. The isoform-specific RI-holoenzyme dimer interface mediated by N3A-N3A' motifs defines multidomain cross-talk and an allosteric network that creates competing roles for ATP and cAMP. Comparisons to the RIIβ holoenzyme demonstrate isoform-specific holoenzyme interfaces and highlights distinct allosteric mechanisms for activation in addition to the structural diversity of the isoforms.
- Published
- 2019
46. Modulating Integrin αIIbβ3 Activity through Mutagenesis of Allosterically Regulated Intersubunit Contacts
- Author
-
Tan, Sophia K, Fong, Karen P, Polizzi, Nicholas F, Sternisha, Alex, Slusky, Joanna SG, Yoon, Kyungchul, DeGrado, William F, and Bennett, Joel S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Alanine ,Allosteric Regulation ,Animals ,CHO Cells ,Cricetinae ,Cricetulus ,Humans ,Mutagenesis ,Platelet Glycoprotein GPIIb-IIIa Complex ,Protein Binding ,Protein Structure ,Secondary ,Protein Structure ,Tertiary ,Protein Subunits ,Medicinal and Biomolecular Chemistry ,Medical Biochemistry and Metabolomics ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical biochemistry and metabolomics ,Medicinal and biomolecular chemistry - Abstract
Integrin αIIbβ3, a transmembrane heterodimer, mediates platelet aggregation when it switches from an inactive to an active ligand-binding conformation following platelet stimulation. Central to regulating αIIbβ3 activity is the interaction between the αIIb and β3 extracellular stalks, which form a tight heterodimer in the inactive state and dissociate in the active state. Here, we demonstrate that alanine replacements of sensitive positions in the heterodimer stalk interface destabilize the inactive conformation sufficiently to cause constitutive αIIbβ3 activation. To determine the structural basis for this effect, we performed a structural bioinformatics analysis and found that perturbing intersubunit contacts with favorable interaction geometry through substitutions to alanine quantitatively accounted for the degree of constitutive αIIbβ3 activation. This mutational study directly assesses the relationship between favorable interaction geometry at mutation-sensitive positions and the functional activity of those mutants, giving rise to a simple model that highlights the importance of interaction geometry in contributing to the stability between protein-protein interactions.
- Published
- 2019
47. Communication between distinct subunit interfaces of the cohesin complex promotes its topological entrapment of DNA.
- Author
-
Guacci, Vincent, Chatterjee, Fiona, Robison, Brett, and Koshland, Douglas
- Subjects
DNA binding ,Mcd1 ,S. cerevisiae ,Scc1 ,Smc3 ,chromosomes ,cohesin ,cohesion ,gene expression ,genetics ,genomics ,Acetylation ,Cell Cycle Proteins ,Chromatids ,Chromosomal Proteins ,Non-Histone ,DNA ,Fungal ,Multiprotein Complexes ,Mutation ,Protein Domains ,Protein Subunits ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins - Abstract
Cohesin mediates higher order chromosome structure. Its biological activities require topological entrapment of DNA within a lumen(s) formed by cohesin subunits. The reversible dissociation of cohesins Smc3p and Mcd1p subunits is postulated to form a regulated gate that allows DNA entry and exit into the lumen. We assessed gate-independent functions of this interface in yeast using a fusion protein that joins Smc3p to Mcd1p. We show that in vivo all the regulators of cohesin promote DNA binding of cohesin by mechanisms independent of opening this gate. Furthermore, we show that this interface has a gate-independent activity essential for cohesin to bind chromosomes. We propose that this interface regulates DNA entrapment by controlling the opening and closing of one or more distal interfaces formed by cohesin subunits, likely by inducing a conformation change in cohesin. Furthermore, cohesin regulators modulate the interface to control both DNA entrapment and cohesin functions after DNA binding.
- Published
- 2019
48. Structural basis for substrate gripping and translocation by the ClpB AAA+ disaggregase.
- Author
-
Rizo, Alexandrea N, Lin, JiaBei, Gates, Stephanie N, Tse, Eric, Bart, Stephen M, Castellano, Laura M, DiMaio, Frank, Shorter, James, and Southworth, Daniel R
- Subjects
Escherichia coli ,Endopeptidase Clp ,Peptides ,Escherichia coli Proteins ,Caseins ,Heat-Shock Proteins ,Protein Subunits ,Adenosine Triphosphate ,Cryoelectron Microscopy ,Protein Transport ,Hydrolysis ,Models ,Molecular ,Protein Aggregates ,AAA Domain ,Models ,Molecular - Abstract
Bacterial ClpB and yeast Hsp104 are homologous Hsp100 protein disaggregases that serve critical functions in proteostasis by solubilizing protein aggregates. Two AAA+ nucleotide binding domains (NBDs) power polypeptide translocation through a central channel comprised of a hexameric spiral of protomers that contact substrate via conserved pore-loop interactions. Here we report cryo-EM structures of a hyperactive ClpB variant bound to the model substrate, casein in the presence of slowly hydrolysable ATPγS, which reveal the translocation mechanism. Distinct substrate-gripping interactions are identified for NBD1 and NBD2 pore loops. A trimer of N-terminal domains define a channel entrance that binds the polypeptide substrate adjacent to the topmost NBD1 contact. NBD conformations at the seam interface reveal how ATP hydrolysis-driven substrate disengagement and re-binding are precisely tuned to drive a directional, stepwise translocation cycle.
- Published
- 2019
49. Analysis of the roles of phosphatidylinositol-4,5-bisphosphate and individual subunits in assembly, localization, and function of Saccharomyces cerevisiae target of rapamycin complex 2
- Author
-
Marshall, Maria Nieves Martinez, Emmerstorfer-Augustin, Anita, Leskoske, Kristin L, Zhang, Lydia H, Li, Biyun, and Thorner, Jeremy
- Subjects
Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Armadillo Domain Proteins ,Carrier Proteins ,Cell Membrane ,Mechanistic Target of Rapamycin Complex 2 ,Phosphatidylinositol 4 ,5-Diphosphate ,Protein Domains ,Protein Subunits ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Eukaryotic cell survival requires maintenance of plasma membrane (PM) homeostasis in response to environmental insults and changes in lipid metabolism. In yeast, a key regulator of PM homeostasis is target of rapamycin (TOR) complex 2 (TORC2), a multiprotein complex containing the evolutionarily conserved TOR protein kinase isoform Tor2. PM localization is essential for TORC2 function. One core TORC2 subunit (Avo1) and two TORC2--associated regulators (Slm1 and Slm2) contain pleckstrin homology (PH) domains that exhibit specificity for binding phosphatidylinositol-4,5-bisphosphate (PtdIns4,5P2). To investigate the roles of PtdIns4,5P2 and constituent subunits of TORC2, we used auxin-inducible degradation to systematically eliminate these factors and then examined localization, association, and function of the remaining TORC2 components. We found that PtdIns4,5P2 depletion significantly reduced TORC2 activity, yet did not prevent PM localization or disassembly of TORC2. Moreover, truncated Avo1 (lacking its C-terminal PH domain) was still recruited to the PM and supported growth. Even when all three PH-containing proteins were absent, the remaining TORC2 subunits were PM-bound. Revealingly, Avo3 localized to the PM independent of both Avo1 and Tor2, whereas both Tor2 and Avo1 required Avo3 for their PM anchoring. Our findings provide new mechanistic information about TORC2 and pinpoint Avo3 as pivotal for TORC2 PM localization and assembly in vivo.
- Published
- 2019
50. Transcription preinitiation complex structure and dynamics provide insight into genetic diseases.
- Author
-
Yan, Chunli, Dodd, Thomas, He, Yuan, Tainer, John A, Tsutakawa, Susan E, and Ivanov, Ivaylo
- Subjects
Humans ,Cell Cycle Proteins ,Transcription Factors ,TFII ,Protein Subunits ,Transcription Factors ,DNA ,Models ,Molecular ,Transcription Factor TFIIH ,Protein Interaction Maps ,Transcription Initiation ,Genetic ,Models ,Molecular ,TFII ,Transcription Initiation ,Genetic ,Chemical Sciences ,Biological Sciences ,Medical and Health Sciences ,Biophysics ,Developmental Biology - Abstract
Transcription preinitiation complexes (PICs) are vital assemblies whose function underlies the expression of protein-encoding genes. Cryo-EM advances have begun to uncover their structural organization. Nevertheless, functional analyses are hindered by incompletely modeled regions. Here we integrate all available cryo-EM data to build a practically complete human PIC structural model. This enables simulations that reveal the assembly's global motions, define PIC partitioning into dynamic communities and delineate how structural modules function together to remodel DNA. We identify key TFIIE-p62 interactions that link core-PIC to TFIIH. p62 rigging interlaces p34, p44 and XPD while capping the DNA-binding and ATP-binding sites of XPD. PIC kinks and locks substrate DNA, creating negative supercoiling within the Pol II cleft to facilitate promoter opening. Mapping disease mutations associated with xeroderma pigmentosum, trichothiodystrophy and Cockayne syndrome onto defined communities reveals clustering into three mechanistic classes that affect TFIIH helicase functions, protein interactions and interface dynamics.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.