4,623 results on '"Department of Biochemistry and Molecular Biophysics"'
Search Results
2. Phytochrome phosphorylation in plant light signaling.
- Author
-
Jeong-Il Kim, Jeoung-Eun Park, Xristo ZaratePresent address: Department of Biochemistry and Molecular Biophysics, University of Arizona, Tucson, AZ 85721, USA., and Pill-Soon Song
- Published
- 2005
3. RRM1 variants cause a mitochondrial DNA maintenance disorder via impaired de novo nucleotide synthesis
- Author
-
Jonathan Shintaku, Wolfgang M. Pernice, Wafaa Eyaid, Jeevan B. GC, Zuben P. Brown, Marti Juanola-Falgarona, Javier Torres-Torronteras, Ewen W. Sommerville, Debby M.E.I. Hellebrekers, Emma L. Blakely, Alan Donaldson, Ingrid van de Laar, Cheng-Shiun Leu, Ramon Marti, Joachim Frank, Kurenai Tanji, David A. Koolen, Richard J. Rodenburg, Patrick F. Chinnery, H.J.M. Smeets, Gráinne S. Gorman, Penelope E. Bonnen, Robert W. Taylor, Michio Hirano, MUMC+: DA KG Lab Specialisten (9), MUMC+: DA KG Lab Centraal Lab (9), RS: MHeNs - R3 - Neuroscience, Klinische Genetica, Institut Català de la Salut, [Shintaku J, Pernice WM, Juanola-Falgarona M] Department of Neurology, H. Houston Merritt Neuromuscular Research Center, Columbia University Irving Medical Center, New York, New York, USA. [Eyaid W] Genetics Division, Department of Pediatrics, King Saud bin Abdulaziz University for Health Science, King Abdulaziz Medical City, Riyadh, Saudi Arabia. [GC JB, Brown ZP] Department of Biochemistry and Molecular Biophysics, Columbia University, New York, New York, USA. [Torres-Torronteras J, Marti R] Center for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain. Grups de Recerca en Malalties Neuromusculars i Mitocondrials, Vall d’Hebron Institut de Recerca (VHIR), Barcelona, Spain. Universitat Autònoma de Barcelona, Bellaterra, Spain, Vall d'Hebron Barcelona Hospital Campus, and Clinical Genetics
- Subjects
DNA Replication ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Mitochondrial Diseases ,Ribonucleoside Diphosphate Reductase ,Nucleotides ,Otros calificadores::Otros calificadores::/genética [Otros calificadores] ,Mitocondris - Malalties - Aspectes genètics ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Nucleosides ,General Medicine ,ADN mitocondrial ,Nutritional and Metabolic Diseases::Metabolic Diseases::Mitochondrial Diseases [DISEASES] ,DNA, Mitochondrial ,DEFICIENCY ,All institutes and research themes of the Radboud University Medical Center ,PHOSPHORYLASE GENE-MUTATIONS ,Nucleic Acids, Nucleotides, and Nucleosides::Nucleic Acids::DNA::DNA, Circular::DNA, Mitochondrial [CHEMICALS AND DRUGS] ,ENCEPHALOMYOPATHY ,nucleótidos y nucleósidos de ácidos nucleicos::ácidos nucleicos::ADN::ADN circular::ADN mitocondrial [COMPUESTOS QUÍMICOS Y DROGAS] ,Ribonucleotide Reductases ,Other subheadings::Other subheadings::/genetics [Other subheadings] ,KINASE ,Humans ,RIBONUCLEOTIDE REDUCTASE ,enfermedades nutricionales y metabólicas::enfermedades metabólicas::enfermedades mitocondriales [ENFERMEDADES] - Abstract
Genetic diseases; Mitochondria; Molecular pathology Enfermedades genéticas; Mitocondrias; Patología molecular Malalties genètiques; Mitocondris; Patologia molecular Mitochondrial DNA (mtDNA) depletion/deletions syndromes (MDDS) encompass a clinically and etiologically heterogenous group of mitochondrial disorders caused by impaired mtDNA maintenance. Among the most frequent causes of MDDS are defects in nucleoside/nucleotide metabolism, which is critical for synthesis and homeostasis of the deoxynucleoside triphosphate (dNTP) substrates of mtDNA replication. A central enzyme for generating dNTPs is ribonucleotide reductase, a critical mediator of de novo nucleotide synthesis composed of catalytic RRM1 subunits in complex with RRM2 or p53R2. Here, we report 5 probands from 4 families who presented with ptosis and ophthalmoplegia as well as other clinical manifestations and multiple mtDNA deletions in muscle. We identified 3 RRM1 loss-of-function variants, including a dominant catalytic site variant (NP_001024.1: p.N427K) and 2 homozygous recessive variants at p.R381, which has evolutionarily conserved interactions with the specificity site. Atomistic molecular dynamics simulations indicate mechanisms by which RRM1 variants affect protein structure. Cultured primary skin fibroblasts of probands manifested mtDNA depletion under cycling conditions, indicating impaired de novo nucleotide synthesis. Fibroblasts also exhibited aberrant nucleoside diphosphate and dNTP pools and mtDNA ribonucleotide incorporation. Our data reveal that primary RRM1 deficiency and, by extension, impaired de novo nucleotide synthesis are causes of MDDS. This work was supported by Department of Defense Focused Program Award W81XWH2010807 (to MH), NIH research grant P01 HD32062 (to MH), and NIH grant 35 GM139453 (to JF). MH is supported by the Arturo Estopinan TK2 Research Fund, Nicholas Nunno Foundation, JDM Fund for Mitochondrial Research, Shuman Mitochondrial Disease Fund, the Marriott Mitochondrial Disease Clinic Research Fund from the J. Willard and Alice S. Marriott Foundation, and NIH grant U54 NS078059. Work in Newcastle upon Tyne was supported by the Wellcome Centre for Mitochondrial Research (203105/Z/16/Z), Medical Research Council International Centre for Genomic Medicine in Neuromuscular Disease (MR/S005021/1), UK NIHR Biomedical Research Centre in Age and Age Related Diseases award to the Newcastle upon Tyne Hospitals NHS Foundation, the Lily Foundation, and the UK National Health Service Highly Specialised Service for Rare Mitochondrial Disorders. RWT receives financial support from the Pathological Society. EWS was funded by a Medical Research Council PhD studentship. This work used the Extreme Science and Engineering Discovery Environment (XSEDE), which is supported by National Science Foundation grant ACI-1548562. JBGC is supported by grant BIO210070 from XSEDE. The authors thank the patients and their families for collaborating in this study and Saba Tadesse for technical support of mitochondrial respiratory chain enzyme activities. We also thank the Genome Technology Center at the Radboud University Medical Center and BGI Copenhagen for WES technical support.
- Published
- 2022
4. Miniprep DNA Isolation for Automated Sequencing of Multiple Samples
- Author
-
Sheibani, Nader and Frazier, William A.
- Published
- 1997
- Full Text
- View/download PDF
5. A single clonal lineage of transmissible cancer identified in two marine mussel species in South America and Europe
- Author
-
Nicolás Merino-Véliz, Florencia Cremonte, Gloria Arriagada, Maryline Houssin, Maurine Hammel, Susan A. Baldwin, Marisa A. Yonemitsu, Annette F. Muttray, Carol L. Reinisch, Stephen P. Goff, Michael J. Metzger, James P. Sherry, Maria Polo-Prieto, Nicolas Bierne, Nuria Natalia Vázquez, Alexis Simon, Rachael M. Giersch, E. A. V. Burioli, Fernando T Avilés, Pacific Northwest Research Institute (PNRI), Howard Hughes Medical Institute [Chevy Chase] (HHMI), Howard Hughes Medical Institute (HHMI), Institut des Sciences de l'Evolution de Montpellier (UMR ISEM), Centre de Coopération Internationale en Recherche Agronomique pour le Développement (Cirad)-École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université de Montpellier (UM)-Institut de recherche pour le développement [IRD] : UR226-Centre National de la Recherche Scientifique (CNRS), Interactions Hôtes-Pathogènes-Environnements (IHPE), Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Institut Français de Recherche pour l'Exploitation de la Mer (IFREMER)-Université de Perpignan Via Domitia (UPVD), Instituto de Biología de Organismos Marinos [Chubut] (IBIOMAR), Consejo Nacional de Investigaciones Científicas y Técnicas [Buenos Aires] (CONICET), Instituto de Ciencias Biomédicas [Santiago, Chile] (Facultad de Medicina), LABÉO, Pôle d’analyses et de recherche de Normandie (LABÉO), Environmental Resources Management, Water Science and Technology, Environment and Climate Change Canada, Chemical and Biological Engineering, University of British Columbia (UBC), Department of Microbiology and Immunology, Columbia University Irving Medical Center (CUIMC), Department of Biochemistry and Molecular Biophysics, Biologie des Organismes et Ecosystèmes Aquatiques (BOREA), Université de Caen Normandie (UNICAEN), Normandie Université (NU)-Normandie Université (NU)-Muséum national d'Histoire naturelle (MNHN)-Institut de Recherche pour le Développement (IRD)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)-Université des Antilles (UA), Departamento de Ciencias Biologicas [Santiago, Chile] (Facultad de Ciencias Biologicas y Facultad de Medicina), Universidad Andrés Bello - UNAB (CHILE), Centre de Coopération Internationale en Recherche Agronomique pour le Développement (Cirad)-École Pratique des Hautes Études (EPHE), Université de Perpignan Via Domitia (UPVD)-Institut Français de Recherche pour l'Exploitation de la Mer (IFREMER)-Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM), Universidad Andrés Bello [Santiago] (UNAB), ANR-18-CE35-0009,TRANSCAN,ECOLOGIE ET EVOLUTION DES CANCERS TRANSMISSIBLES(2018), École pratique des hautes études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université de Montpellier (UM)-Centre de Coopération Internationale en Recherche Agronomique pour le Développement (Cirad)-Centre National de la Recherche Scientifique (CNRS)-Institut de recherche pour le développement [IRD] : UR226, Centre National de la Recherche Scientifique (CNRS)-Université des Antilles (UA)-Muséum national d'Histoire naturelle (MNHN)-Institut de Recherche pour le Développement (IRD)-Sorbonne Université (SU)-Université de Caen Normandie (UNICAEN), and Normandie Université (NU)-Normandie Université (NU)
- Subjects
0106 biological sciences ,0301 basic medicine ,Mytilus chilensis ,Aquatic Organisms ,Mytilus edulis ,DISEASES TRANSMISIBLES ,01 natural sciences ,ARGENTINE SEA ,purl.org/becyt/ford/1 [https] ,Neoplasms ,2.1 Biological and endogenous factors ,Biology (General) ,Aetiology ,Phylogeny ,Cancer Biology ,cancer biology ,Microbiology and Infectious Disease ,Phylogenetic tree ,General Neuroscience ,[SDV.BID.EVO]Life Sciences [q-bio]/Biodiversity/Populations and Evolution [q-bio.PE] ,General Medicine ,Single Nucleotide ,CANCER ,BIVALVES ,Mytilus ,3. Good health ,Europe ,Medicine ,transmissible cancer ,Sequence Analysis ,CIENCIAS NATURALES Y EXACTAS ,Infectious agent ,Research Article ,QH301-705.5 ,Science ,infectious disease ,Zoology ,Single-nucleotide polymorphism ,Biology ,010603 evolutionary biology ,Polymorphism, Single Nucleotide ,General Biochemistry, Genetics and Molecular Biology ,bivalve ,Ciencias Biológicas ,03 medical and health sciences ,bivalve transmissible neoplasia ,Genetics ,Animals ,14. Life underwater ,Allele ,Polymorphism ,purl.org/becyt/ford/1.6 [https] ,Alleles ,General Immunology and Microbiology ,microbiology ,Mussel ,Sequence Analysis, DNA ,DNA ,South America ,Biología Marina, Limnología ,biology.organism_classification ,030104 developmental biology ,Infectious disease (medical specialty) ,Other ,Biochemistry and Cell Biology - Abstract
Transmissible cancers, in which cancer cells themselves act as an infectious agent, have been identified in Tasmanian devils, dogs, and four bivalves. We investigated a disseminated neoplasia affecting geographically distant populations of two species of mussels (Mytilus chilensis in South America and M. edulis in Europe). Sequencing alleles from four loci (two nuclear and two mitochondrial) provided evidence of transmissible cancer in both species. Phylogenetic analysis of cancer-associated alleles and analysis of diagnostic SNPs showed that cancers in both species likely arose in a third species of mussel (M. trossulus), but these cancer cells are independent from the previously identified transmissible cancer in M. trossulus from Canada. Unexpectedly, cancers from M. chilensis and M. edulis are nearly identical, showing that the same cancer lineage affects both. Thus, a single transmissible cancer lineage has crossed into two new host species and has been transferred across the Atlantic and Pacific Oceans and between the Northern and Southern hemispheres., eLife digest Cancer cells can grow and spread in one individual, but they normally do not spread to others. There are a few exceptions to this rule. For example, there are cancers in Tasmanian devils, dogs and bivalve shellfish that can spread to other members of the same species. In these creatures, cancer from one individual evolved the ability to spread throughout the population. These cancer cells infect animals like a pathogen. A fatal cancer called disseminated neoplasia affects many species of bivalves. In four bivalve species, including the marine mussel Mytilus trossulus, scientists have shown that the cancer can spread from one individual to another. This transmissible cancer has been found in M. trossulus mussels in British Columbia, Canada; but related species of mussels in other parts of the world also develop disseminated neoplasia. It is possible these other cancers are transmissible and have spread from one population of mussels to another. Yonemitsu et al. performed genetic analyses to show that cancers found in two other mussel species – Mytilus chilensis in South America and Mytilus edulis in Europe – are transmissible and arose in M. trossulus. The cancers in the South American and European mussels were nearly identical genetically, which suggests that they came from a single M. trossulus mussel with cancer at some point in the past. Somehow cancer cells spread between the Northern and the Southern Hemispheres and across the Atlantic Ocean, infecting multiple species across the world. The analyses also show that this cancer lineage is different from the one previously identified in British Columbia. These analyses show that bivalve transmissible neoplasia was able to spread worldwide, most likely through accidental transport of infected mussels on international shipping vessels. This suggests that human activities unwittingly introduced the disease to new areas. Learning more about transmissible cancers may help scientists understand how cancers evolve with their hosts in extreme situations.
- Published
- 2019
- Full Text
- View/download PDF
6. Use of plasmon waveguide resonance (PWR) spectroscopy for examining binding, signaling and lipid domain partitioning of membrane proteins
- Author
-
Isabel D. Alves, Scott Cowell, Gordon Tollin, Zdzislaw Salamon, Victor J. Hruby, Université Pierre et Marie Curie - Paris 6 (UPMC), Department of Biochemistry and Molecular Biophysics, and University of Arizona
- Subjects
Cell signaling ,Lipid Bilayers ,CHO Cells ,Ligands ,General Biochemistry, Genetics and Molecular Biology ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,Cricetulus ,Growth factor receptor ,Cricetinae ,Receptors, Opioid, delta ,Animals ,Humans ,Inverse agonist ,General Pharmacology, Toxicology and Pharmaceutics ,Integral membrane protein ,Lipid raft ,Ion channel ,030304 developmental biology ,G protein-coupled receptor ,0303 health sciences ,[CHIM.ORGA]Chemical Sciences/Organic chemistry ,Chemistry ,Spectrum Analysis ,030302 biochemistry & molecular biology ,Membrane Proteins ,General Medicine ,Surface Plasmon Resonance ,Membrane protein ,Biochemistry ,Biophysics ,Protein Binding ,Signal Transduction - Abstract
International audience; AIMS: Due to their anisotropic properties and other factors, it has been difficult to determine the conformational and dynamic properties of integral membrane proteins such as G-protein coupled receptors (GPCRs), growth factor receptors, ion channels, etc. in response to ligands and subsequent signaling. Herein a novel methodology is presented that allows such studies to be performed while maintaining the receptors in a membrane environment. MAIN METHOD: Plasmon waveguide resonance (PWR) spectroscopy is a relatively new biophysical method which allows one to directly observe structural and dynamic changes which occur on interaction of GPCRs (and other integral membrane proteins) with ligands and signaling molecules. The delta opioid receptor (DOR) and its ligands serve as an excellent model system to illustrate the new insights into GPCR signaling that can be obtained by this method. KEY FINDINGS: Among our key findings are: 1) it is possible to obtain the following information directly and without any need for labels (radioactive, fluorescent, etc.): binding affinities, and the ability to distinguish between agonists, antagonists, inverse agonist, and partial agonists without a need for second messenger analysis; 2) it is possible to determine directly, again without a need for labels, G-protein binding to variously occupied or unoccupied DORs, and to determine which alpha-subtype is involved in allowing structurally different agonist ligands to have differential effects; 3) GTPgammaS binding can be examined directly; and 4) binding of the DOR with different ligands leads to differential segregation of the ligand-receptor complex into lipid rafts. SIGNIFICANCE: The implications of these discoveries suggest a need to modify our current views of GPCR-ligand interactions and signaling.
- Published
- 2010
- Full Text
- View/download PDF
7. HDAC6 activity is a non-oncogene addiction hub for inflammatory breast cancers
- Author
-
Mary L. Alpaugh, Laura P. Saucedo-Cuevas, Pascal Finetti, Min Yang, David Llobet-Navas, Andrea Califano, Veronica Castro, Wendy A. Woodward, Jiyang Yu, François Bertucci, Eva Murga-Penas, Patricia Villagrasa, Daniel Birnbaum, Preeti Putcha, Steven N. Quayle, Ruth Rodriguez-Barrueco, Jose M. Silva, Herbert Irving Comprehensive Cancer Center, Columbia University [New York], Departments of Biomedical Informatics & Systems Biolog, Columbia University [New York]-Columbia University Irving Medical Center (CUIMC), Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center [Houston], Department of Pathology, Icahn School of Medicine at Mount Sinai [New York] (MSSM), Columbia University Irving Medical Center (CUIMC), Acetylon Pharmaceuticals, Inc., Centre de Recherche en Cancérologie de Marseille (CRCM), Aix Marseille Université (AMU)-Institut Paoli-Calmettes, Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Fédération nationale des Centres de lutte contre le Cancer (FNCLCC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Department of Surgery, Memorial Sloane Kettering Cancer Center [New York], Department of Biochemistry and Molecular Biophysics, BMC, BMC, and Pathology/molecular and cellular medicine
- Subjects
Medicine(all) ,business.industry ,medicine.medical_treatment ,[SDV]Life Sciences [q-bio] ,HDAC6 ,medicine.disease ,Oncogene Addiction ,Phenotype ,Inflammatory breast cancer ,3. Good health ,Targeted therapy ,[SDV] Life Sciences [q-bio] ,Breast cancer ,Surgical oncology ,RNA interference ,medicine ,Cancer research ,business ,skin and connective tissue diseases - Abstract
International audience; AbstractIntroductionInflammatory breast cancer (IBC) is the most lethal form of breast cancers with a 5-year survival rate of only 40 %. Despite its lethality, IBC remains poorly understood which has greatly limited its therapeutic management. We thus decided to utilize an integrative functional genomic strategy to identify the Achilles’ heel of IBC cells.MethodsWe have pioneered the development of genetic tools as well as experimental and analytical strategies to perform RNAi-based loss-of-function studies at a genome-wide level. Importantly, we and others have demonstrated that these functional screens are able to identify essential functions linked to certain cancer phenotypes. Thus, we decided to use this approach to identify IBC specific sensitivities.ResultsWe identified and validated HDAC6 as a functionally necessary gene to maintain IBC cell viability, while being non-essential for other breast cancer subtypes. Importantly, small molecule inhibitors for HDAC6 already exist and are in clinical trials for other tumor types. We thus demonstrated that Ricolinostat (ACY1215), a leading HDAC6 inhibitor, efficiently controls IBC cell proliferation both in vitro and in vivo. Critically, functional HDAC6 dependency is not associated with genomic alterations at its locus and thus represents a non-oncogene addiction. Despite HDAC6 not being overexpressed, we found that its activity is significantly higher in IBC compared to non-IBC cells, suggesting a possible rationale supporting the observed dependency.ConclusionOur finding that IBC cells are sensitive to HDAC6 inhibition provides a foundation to rapidly develop novel, efficient, and well-tolerated targeted therapy strategies for IBC patients.
- Published
- 2015
- Full Text
- View/download PDF
8. Evidence for functional redundancy between C. elegans ADAM proteins SUP-17/Kuzbanian and ADM-4/TACE
- Author
-
Sophie Jarriault, Iva Greenwald, and Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
- Subjects
Male ,ADAM10 ,[SDV]Life Sciences [q-bio] ,Kuzbanian ,0302 clinical medicine ,[SDV.BC.IC]Life Sciences [q-bio]/Cellular Biology/Cell Behavior [q-bio.CB] ,Receptor ,Caenorhabditis elegans ,Phylogeny ,ComputingMilieux_MISCELLANEOUS ,Genetics ,0303 health sciences ,Metalloproteinase ,SUP-17 ,Membrane Glycoproteins ,biology ,Receptors, Notch ,Metalloendopeptidases ,ADAM Proteins ,Phenotype ,Zinc ,Ectodomain ,Notch proteins ,C. elegans ,Female ,Notch ,ADAM17 Protein ,Article ,Metalloprotease ,03 medical and health sciences ,Animals ,Reprolysin ,Caenorhabditis elegans Proteins ,Spermatogenesis ,Molecular Biology ,030304 developmental biology ,TACE ,ADAM ,Membrane Proteins ,ADM-4 ,Cell Biology ,biology.organism_classification ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,LIN-12 ,Mutation ,Oocytes ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The ectodomain of LIN-12/Notch proteins is cleaved and shed upon ligand binding. In C. elegans, genetic evidence has implicated SUP-17, the ortholog of Drosophila Kuzbanian and mammalian ADAM10, as the protease that mediates this event. In mammals, however, biochemical evidence has implicated TACE, a different ADAM protein. We have investigated potential functional redundancy of sup-17 and the C. elegans ortholog of TACE, adm-4, by exploring their roles in cell fate decisions mediated by lin-12/Notch genes. We found that reduced adm-4 activity, like reduced sup-17 activity, suppresses an allele of glp-1 that encodes a constitutively active receptor. Furthermore, concomitant reduction of adm-4 and sup-17 activity causes the production of two anchor cells in the hermaphrodite gonad, instead of one--a phenotype associated with loss of lin-12 activity. We also identified a novel fertility defect in the somatic gonad when both sup-17 and adm-4 are depleted. Expression of a truncated form of LIN-12 that mimics the product of ectodomain shedding rescues this fertility defect, suggesting that sup-17 and adm-4 may mediate ectodomain shedding of LIN-12 and/or GLP-1. Our results are consistent with the possibility that sup-17 and adm-4 are functionally redundant for at least a subset of LIN-12/Notch mediated decisions in C. elegans.
- Published
- 2005
- Full Text
- View/download PDF
9. Branched amphiphilic cationic oligopeptides form peptiplexes with DNA: a study of their biophysical properties and transfection efficiency
- Author
-
Robert Szoszkiewicz, Sushanth Gudlur, Stella Y. Lee, Luís Carlos de Souza Ferreira, Takeo Iwamoto, Pinakin Sukthankar, Ladislav Šimo, L. Adriana Avila, John M. Tomich, Nicoleta Ploscariu, Luana R.M.M. Aps, Yoonseong Park, Department of Biochemistry and Molecular Biophysics (DBMB), Columbia University [New York], Institute of Biomedical Sciences, Universidad de Chile = University of Chile [Santiago] (UCHILE), Department of Physics, Tamkang University [New Taipei] (TKU), Department of Entomology, Michigan State University [East Lansing], Michigan State University System-Michigan State University System, Division of Biology, Kansas State University, Division of Biochemistry - Core Research Facilities, The Jikei University School of Medicine, PHS-NIH Grant R01 GM074096, Terry Johnson Cancer Center at Kansas State University, and JSPS KAKENHI 23590649
- Subjects
nontoxic ,Cell Survival ,Macromolecular Substances ,gene deliver ,[SDV]Life Sciences [q-bio] ,Pharmaceutical Science ,Peptide ,Gene delivery ,Transfection ,nonviral ,Biophysical Phenomena ,HeLa ,Surface-Active Agents ,chemistry.chemical_compound ,Plasmid ,plasmid DNA ,cationic branched peptides ,Cations ,nanofibers ,Drug Discovery ,Amphiphile ,Humans ,chemistry.chemical_classification ,Oligopeptide ,biology ,Gene Transfer Techniques ,condensed DNA ,DNA ,Genetic Therapy ,biology.organism_classification ,Nanostructures ,chemistry ,Biochemistry ,peptiplex ,BAPCs ,Molecular Medicine ,Oligopeptides ,HeLa Cells - Abstract
Over the past decade, peptides have emerged as a new family of potential carriers in gene therapy. Peptides are easy to synthesize and quite stable. Additionally, sequences shared by the host proteome are not expected to be immunogenic or trigger inflammatory responses, which are commonly observed with viral approaches. We recently reported on a new class of branched amphiphilic peptide capsules (BAPCs) that self-assemble into extremely stable nanospheres. These capsules are capable of retaining and delivering alpha-emitting radionuclides to cells. Here we report that, in the presence of double stranded plasmid DNA, BAPCs are unable to form. Instead, depending of the peptide/DNA ratios, the peptides either coat the plasmid surface forming nanofibers (high peptide to DNA ratio) or condense the plasmid into nanometer-sized compacted structures (at low peptide to DNA ratios). Different gene delivery efficiencies are observed for the two types of assemblies. The compacted nanometer-sized structures display much higher transfection efficiencies in HeLa cells. This level of transfection is greater than that observed for a lipid-based reagent when the total number of viable transfected cells is taken into account.
- Published
- 2015
- Full Text
- View/download PDF
10. Structural basis for catalysis in a CDP-alcohol phosphotransferase
- Author
-
Giuliano Sciara, Joseph H. Graziano, David Tomasek, Kanagalaghatta R. Rajashankar, Lawrence Shapiro, Vesna Slavkovic, Oliver B. Clarke, Filippo Mancia, Rushelle Byfield, Shantelle Tabuso, Surajit Banerjee, Raphael Cohn, Brian Kloss, Biodiversité et Biotechnologie Fongiques (BBF), École Centrale de Marseille (ECM)-Aix Marseille Université (AMU)-Institut National de la Recherche Agronomique (INRA), Department of Physiology and Cellular Biophysics, Columbia University College of Physicians and Surgeons, Department of Biochemistry and Molecular Biophysics (DBMB), Columbia University [New York], New York Structural Biology Center (NYSBC), Rockefeller University [New York]-Columbia University [New York]-New York University [New York] (NYU), NYU System (NYU)-NYU System (NYU)-City University of New York [New York] (CUNY)-Memorial Sloane Kettering Cancer Center [New York]-Wadsworth Center, New York State Department of Health [Albany]-New York State Department of Health [Albany]-Weill Medical College of Cornell University [New York]- Albert Einstein College of Medicine-Icahn School of Medicine at Mount Sinai [New York] (MSSM), Department of Chemistry and Chemical Biology, Rutgers Sate University, Department of Environmental Health Sciences, Mailman School of Public Health, Institut National de la Recherche Agronomique (INRA)-Aix Marseille Université (AMU)-École Centrale de Marseille (ECM), Columbia University [New York]-Wadsworth Center, New York State Department of Health [Albany]-New York State Department of Health [Albany]-New York University [New York] (NYU), NYU System (NYU)-NYU System (NYU)-City University of New York [New York] (CUNY)-Rockefeller University [New York]-Memorial Sloane Kettering Cancer Center [New York]-Icahn School of Medicine at Mount Sinai [New York] (MSSM)- Albert Einstein College of Medicine [New York]-Weill Medical College of Cornell University [New York], Rutgers University [Camden], Rutgers University System (Rutgers)-Rutgers University System (Rutgers), and Mancia, Filippo
- Subjects
Models, Molecular ,Stereochemistry ,Archaeal Proteins ,[SDV]Life Sciences [q-bio] ,Amino Acid Motifs ,Molecular Sequence Data ,General Physics and Astronomy ,Protomer ,Bioinformatics ,General Biochemistry, Genetics and Molecular Biology ,Article ,phosphotransférase system ,Phosphotransferase ,03 medical and health sciences ,Protein structure ,Catalytic Domain ,Transferase ,Amino Acid Sequence ,phospholipide membranaire ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,Binding Sites ,biology ,030302 biochemistry & molecular biology ,Archaeoglobus fulgidus ,Active site ,General Chemistry ,PEP group translocation ,Protein Structure, Tertiary ,carbohydrates (lipids) ,Transmembrane domain ,Phosphotransferases (Alcohol Group Acceptor) ,Alcohols ,biology.protein ,Biocatalysis ,lipids (amino acids, peptides, and proteins) ,escherichia coli ,Sequence Alignment - Abstract
The CDP-alcohol phosphotransferase (CDP-AP) family of integral membrane enzymes catalyses the transfer of a substituted phosphate group from a CDP-linked donor to an alcohol acceptor. This is an essential reaction for phospholipid biosynthesis across all kingdoms of life, and it is catalysed solely by CDP-APs. Here we report the 2.0 Å resolution crystal structure of a representative CDP-AP from Archaeoglobus fulgidus. The enzyme (AF2299) is a homodimer, with each protomer consisting of six transmembrane helices and an N-terminal cytosolic domain. A polar cavity within the membrane accommodates the active site, lined with the residues from an absolutely conserved CDP-AP signature motif (D(1)xxD(2)G(1)xxAR...G(2)xxxD(3)xxxD(4)). Structures in the apo, CMP-bound, CDP-bound and CDP-glycerol-bound states define functional roles for each of these eight conserved residues and allow us to propose a sequential, base-catalysed mechanism universal for CDP-APs, in which the fourth aspartate (D4) acts as the catalytic base.
- Published
- 2014
- Full Text
- View/download PDF
11. Temporal patterning of Drosophila medulla neuroblasts controls neural fates
- Author
-
Claude Desplan, Ted Erclik, Roumen Voutev, Xin Li, Javier Morante, Zhenqing Chen, Arzu Celik, Claire Bertet, Srinidhi Venkatesh, School of Materials Science and Engineering, University of New South Wales [Sydney] (UNSW), New York University, New York University [New York] (NYU), NYU System (NYU)-NYU System (NYU), Institut de Biologie du Développement de Marseille (IBDM), Aix Marseille Université (AMU)-Centre National de la Recherche Scientifique (CNRS), Department of Biochemistry and Molecular Biophysics (DBMB), Columbia University [New York], Institute for Genetics and Center for Molecular Medicine, University of Cologne, and Department of Biology, New York University, 1009 Silver Center
- Subjects
Male ,animal structures ,Time Factors ,Cellular differentiation ,[SDV]Life Sciences [q-bio] ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Visual system ,Article ,03 medical and health sciences ,0302 clinical medicine ,Neuroblast ,Neural Stem Cells ,Animals ,Drosophila Proteins ,Cell Lineage ,Visual Pathways ,Transcription factor ,[SDV.BDD]Life Sciences [q-bio]/Development Biology ,Medulla ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Neurons ,0303 health sciences ,Multidisciplinary ,fungi ,Brain ,Cell Differentiation ,[SDV.BDD.MOR]Life Sciences [q-bio]/Development Biology/Morphogenesis ,Anatomy ,biology.organism_classification ,Neural stem cell ,Drosophila melanogaster ,Gene Expression Regulation ,Female ,Neuroscience ,030217 neurology & neurosurgery ,Drosophila Protein ,Transcription Factors - Abstract
In the Drosophila optic lobes, the medulla processes visual information coming from inner photoreceptors R7 and R8 and from lamina neurons. It contains approximately 40,000 neurons belonging to more than 70 different types. Here we describe how precise temporal patterning of neural progenitors generates these different neural types. Five transcription factors-Homothorax, Eyeless, Sloppy paired, Dichaete and Tailless-are sequentially expressed in a temporal cascade in each of the medulla neuroblasts as they age. Loss of Eyeless, Sloppy paired or Dichaete blocks further progression of the temporal sequence. We provide evidence that this temporal sequence in neuroblasts, together with Notch-dependent binary fate choice, controls the diversification of the neuronal progeny. Although a temporal sequence of transcription factors had been identified in Drosophila embryonic neuroblasts, our work illustrates the generality of this strategy, with different sequences of transcription factors being used in different contexts.
- Published
- 2013
- Full Text
- View/download PDF
12. Divergent transcriptional regulatory logic at the intersection of tissue growth and developmental patterning
- Author
-
Lijia Ma, Kevin P. White, Richard S. Mann, Matthew Slattery, Roumen Voutev, Nicolas Nègre, Department of Biochemistry and Molecular Biophysics (DBMB), Columbia University [New York], Institute for Genomics and Systems Biology - Department of Human Genetics, University of Chicago, Diversité, Génomes & Interactions Microorganismes - Insectes [Montpellier] (DGIMI), Université de Montpellier (UM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Institut National de la Recherche Agronomique (INRA), NIH [GM058575, U01HG004264, GM087047], Leukemia and Lymphoma Society CDP fellowship, and Institut National de la Recherche Agronomique (INRA)-Université Montpellier 2 - Sciences et Techniques (UM2)-Université de Montpellier (UM)
- Subjects
Developmental genetics ,Cancer Research ,lcsh:QH426-470 ,Transcription, Genetic ,[SDV]Life Sciences [q-bio] ,Biology ,Eye ,Tissues--Growth ,03 medical and health sciences ,0302 clinical medicine ,Genetic transcription--Regulation ,Developmental biology ,Coactivator ,Genetics ,Animals ,Drosophila Proteins ,Wings, Animal ,Enhancer ,Molecular Biology ,Transcription factor ,Genetics (clinical) ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Regulation of gene expression ,Homeodomain Proteins ,0303 health sciences ,Binding Sites ,fungi ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,YAP-Signaling Proteins ,Chromatin ,DNA binding site ,DNA-Binding Proteins ,lcsh:Genetics ,Drosophila melanogaster ,Organ Specificity ,FOS: Biological sciences ,Trans-Activators ,Chromatin immunoprecipitation ,030217 neurology & neurosurgery ,Binding domain ,Research Article ,Signal Transduction ,Transcription Factors - Abstract
The Yorkie/Yap transcriptional coactivator is a well-known regulator of cellular proliferation in both invertebrates and mammals. As a coactivator, Yorkie (Yki) lacks a DNA binding domain and must partner with sequence-specific DNA binding proteins in the nucleus to regulate gene expression; in Drosophila, the developmental regulators Scalloped (Sd) and Homothorax (Hth) are two such partners. To determine the range of target genes regulated by these three transcription factors, we performed genome-wide chromatin immunoprecipitation experiments for each factor in both the wing and eye-antenna imaginal discs. Strong, tissue-specific binding patterns are observed for Sd and Hth, while Yki binding is remarkably similar across both tissues. Binding events common to the eye and wing are also present for Sd and Hth; these are associated with genes regulating cell proliferation and “housekeeping” functions, and account for the majority of Yki binding. In contrast, tissue-specific binding events for Sd and Hth significantly overlap enhancers that are active in the given tissue, are enriched in Sd and Hth DNA binding sites, respectively, and are associated with genes that are consistent with each factor's previously established tissue-specific functions. Tissue-specific binding events are also significantly associated with Polycomb targeted chromatin domains. To provide mechanistic insights into tissue-specific regulation, we identify and characterize eye and wing enhancers of the Yki-targeted bantam microRNA gene and demonstrate that they are dependent on direct binding by Hth and Sd, respectively. Overall these results suggest that both Sd and Hth use distinct strategies – one shared between tissues and associated with Yki, the other tissue-specific, generally Yki-independent and associated with developmental patterning – to regulate distinct gene sets during development., Author Summary The Hippo tumor suppressor pathway controls proliferation in a tissue-nonspecific fashion in Drosophila epithelial progenitor tissues via the transcriptional coactivator Yorkie (Yki). However, despite the tissue-nonspecific role that Yki plays in tissue growth, the transcription factors that recruit Yki to DNA, most notably Scalloped (Sd) and Homothorax (Hth), are important regulators of developmental patterning with many tissue-specific functions. Thus, these three transcriptional regulators – Yki, Sd, and Hth – provide a model for exploring the properties of protein-DNA interactions that regulate both tissue-shared and tissue-specific functions. With this goal in mind, we identified the positions in the fly genome that are bound by Yki, Sd, and Hth in the progenitors of the wing and eye-antenna structures of the fly. These data not only provide a global view of the Yki gene regulatory network, they reveal an unusual amount of tissue specificity in the genomic regions targeted by Sd and Hth, but not Yki. The data also reveal that tissue-specific binding is very likely to overlap tissue-specific enhancer regions, provide important clues for how tissue-specific Sd and Hth binding occurs, and support the idea that gene regulatory networks are plastic, with spatial differences in binding significantly impacting network structures.
- Published
- 2013
- Full Text
- View/download PDF
13. Functional analyses of variants reveal a significant role for dominant negative and common alleles in oligogenic Bardet–Biedl syndrome
- Author
-
Norann A. Zaghloul, Edwin C. Oh, Jantje M. Gerdes, Cecilia Gascue, Jose L. Badano, Rudolph L. Leibel, Jonathan Binkley, Yangjian Liu, Carmen C. Leitch, Nicholas Katsanis, Arend Sidow, Yana Bromberg, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Institut Pasteur de Montevideo, Réseau International des Instituts Pasteur (RIIP), Center for Human Disease Modeling, Duke University [Durham], Johns Hopkins University (JHU), Department of Biochemistry and Molecular Biophysics, Columbia University [New York], Columbia University Center for Computational Biology and Bioinformatics, Department of Genetics [Stanford], Stanford Medicine, Stanford University-Stanford University, Department of Pathology [Stanford], and Division of Molecular Genetics and Naomi Berrie Diabetes Center
- Subjects
Male ,MESH: Mutation ,MESH: Pedigree ,Biology ,MESH: Phenotype ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,MESH: Bardet-Biedl Syndrome ,Bardet–Biedl syndrome ,Genetic variation ,medicine ,Animals ,Humans ,MESH: Animals ,Allele ,MESH: Zebrafish ,Bardet-Biedl Syndrome ,Alleles ,Zebrafish ,030304 developmental biology ,Genetics ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,0303 health sciences ,Mutation ,MESH: Humans ,Multidisciplinary ,MESH: Alleles ,Biological Sciences ,medicine.disease ,MESH: Gene Expression Regulation ,MESH: Male ,Human genetics ,Pedigree ,Genetic load ,Ciliopathy ,Phenotype ,MESH: Models, Animal ,Gene Expression Regulation ,Models, Animal ,Epistasis ,Female ,MESH: Female ,030217 neurology & neurosurgery - Abstract
Technological advances hold the promise of rapidly catalyzing the discovery of pathogenic variants for genetic disease. However, this possibility is tempered by limitations in interpreting the functional consequences of genetic variation at candidate loci. Here, we present a systematic approach, grounded on physiologically relevant assays, to evaluate the mutational content (125 alleles) of the 14 genes associated with Bardet–Biedl syndrome (BBS). A combination of in vivo assays with subsequent in vitro validation suggests that a significant fraction of BBS-associated mutations have a dominant-negative mode of action. Moreover, we find that a subset of common alleles, previously considered to be benign, are, in fact, detrimental to protein function and can interact with strong rare alleles to modulate disease presentation. These data represent a comprehensive evaluation of genetic load in a multilocus disease. Importantly, superimposition of these results to human genetics data suggests a previously underappreciated complexity in disease architecture that might be shared among diverse clinical phenotypes.
- Published
- 2010
- Full Text
- View/download PDF
14. Diversity of Chemical Mechanisms in Thioredoxin Catalysis Revealed by Single-Molecule Force Spectroscopy
- Author
-
Eric A. Gaucher, Julio M. Fernandez, Seung-Hyun Cho, Inmaculada Sanchez-Romero, Bruce J. Berne, Eric Gelhaye, Pallav Kosuri, Jose M. Sanchez-Ruiz, Ana Chueca, Katja Becker, Arun P. Wiita, Raul Perez-Jimenez, Jean P Jacquot, Antonio Miranda-Vizuete, Jingyuan Li, David Rodriguez-Larrea, Jon Beckwith, Arne Holmgren, Columbia University [New York], Department of Chemistry, Department of Biochemistry and Molecular Biophysics (DBMB), Universidad de Granada (UGR), Estación Experimental del Zaidín (EEZ), Consejo Superior de Investigaciones Científicas [Madrid] (CSIC), Karolinska Institutet [Stockholm], Universidad Pablo de Olavide, Justus-Liebig-Universität Gießen (JLU), Department of Microbiology and Molecular Genetics, Harvard Medical School [Boston] (HMS), Interactions Arbres-Microorganismes (IAM), Institut National de la Recherche Agronomique (INRA)-Université de Lorraine (UL), School of Biology, and Georgia Institute of Technology [Atlanta]
- Subjects
ESCHERICHIA-COLI THIOREDOXIN ,Models, Molecular ,Magnetic Resonance Spectroscopy ,[SDV]Life Sciences [q-bio] ,Crystallography, X-Ray ,01 natural sciences ,Thioredoxins ,Structural Biology ,Bacterial proteins ,CRYSTAL-STRUCTURES ,Disulfides ,Phylogeny ,chemistry.chemical_classification ,0303 health sciences ,PLASMODIUM-FALCIPARUM ,3. Good health ,Isoenzymes ,ASCORBATE OXIDASE ,Eukaryotic Cells ,Biochemistry ,REDOX POTENTIALS ,Protein folding ,Thioredoxin ,Oxidoreductases ,Macromolecule ,animal structures ,Molecular Sequence Data ,Protein degradation ,010402 general chemistry ,REDUCED FORM ,CHLAMYDOMONAS-REINHARDTII ,Article ,Catalysis ,Evolution, Molecular ,03 medical and health sciences ,Species Specificity ,Oxidoreductase ,Molecule ,Animals ,Humans ,Computer Simulation ,Amino Acid Sequence ,Binding site ,Molecular Biology ,030304 developmental biology ,Binding Sites ,Sequence Homology, Amino Acid ,Force spectroscopy ,Genetic Variation ,0104 chemical sciences ,Protein Structure, Tertiary ,Kinetics ,chemistry ,DROSOPHILA-MELANOGASTER ,Biophysics ,ARABIDOPSIS-THALIANA ,INTRAMOLECULAR ELECTRON-TRANSFER - Abstract
22 páginas, 5 figuras, 1 tabla. Material suplementario 10 páginas, 2 figuras., Thioredoxins (Trxs) are oxidoreductase enzymes, present in all organisms, that catalyze the reduction of disulfide bonds in proteins. By applying a calibrated force to a substrate disulfide, the chemical mechanisms of Trx catalysis can be examined in detail at the single-molecule level. Here we use single-molecule force-clamp spectroscopy to explore the chemical evolution of Trx catalysis by probing the chemistry of eight different Trx enzymes. All Trxs show a characteristic Michaelis-Menten mechanism that is detected when the disulfide bond is stretched at low forces, but at high forces, two different chemical behaviors distinguish bacterial-origin from eukaryotic-origin Trxs. Eukaryotic-origin Trxs reduce disulfide bonds through a single-electron transfer reaction (SET), whereas bacterial-origin Trxs show both nucleophilic substitution (SN2) and SET reactions. A computational analysis of Trx structures identifies the evolution of the binding groove as an important factor controlling the chemistry of Trx catalysis., This work was supported by the US National Institutes of Health grants HL66030 and HL61228 to J.M.F., grant GMO55090 to J.B., grant GM43340 to B.J.B., grant I.C.E. (Columbia University) to B.J.B. and J.M.F., grant BIO2006-07332 from the Spanish Ministry of Education and Science and FEDER Funds to J.M.S.-R.
- Published
- 2009
- Full Text
- View/download PDF
15. A global benchmark study using affinity-based biosensors
- Author
-
Aykut Üren, Stephen G. Brohawn, Eva Muñoz, Kenneth Miller, Mike Scott, Heather Hughes, Yasmina Noubia Abdiche, Rohn Lee Millican, Anne W Emerick, Jonathan Brooks, Michael B. Murphy, Charlene S. Lee, Carmelo Di Primo, Joshua Ballard, Quyhn Trinh, Giuseppe A. Papalia, Cynthia S. Hinck, Kevin Thompson, Peter Flynn, Joshua S. Klein, Martin A. Wear, Mark Toews, Richard N. Bohnsack, Mohammed Yousef, Daniel Malashock, Gaetano Barbato, Jill Raymond, Alison Joyce, Min Hsiang Yang, Julie Rothacker, Patrick England, Ashique Rafique, Jacinto López-Sagaseta, Scott L. Klakamp, Daulet Satpaev, Monica Dines, Danny Terwey, Islay Campbell, Joshua Thompson, Ben Busby, Tanmoy Ganguly, Gerardo Gutierrez-Sanchez, Sergei Shikov, Kevin Lindquist, Momchilo Vuyisich, Asya Grinberg, Krista Witte, Yun Hee Cho, Yue Ji Li, Hubert Mantz, Ewa Pol, Gonzalo Obal, Brian J. Pak, María J. Hernáiz, Peter Kainz, Erk Gedig, Mary M. Murphy, Tsafir Bravman, Janus Krarup, Gabrielle Zeder-Lutz, John Quinn, Thomas E. Ryan, Pietro Brandani, Dawn Kernaghan, Anca Clabbers, M. Brent Waddell, Agnes Puskas, Petr Skládal, Sanjay Nilapwar, Kara Herlihy, Gregor Anderluh, Federico Torta, Eric Hommema, Andrej Bavdek, Rejane Guimaraes, Anthony M. Giannetti, Eric Fang, Henrik Anderson, Anne Birgitte Bagge Hagel, Schuyler B Corry, Mark R. Witmer, Phillippe Neuner, Sébastien Wieckowski, Nico Dankbar, Alanna Pinkerton, Suparna Mundodo, Trevor D. Chapman, Sylvie Canepa, Satya P. Yadav, Rostislav Skrabana, Oleksandr Kalyuzhniy, Phini S Katsamba, Vidya Chandrasekaran, Eugene G. Chomey, Dana Reichmann, Katy McGirr, Dotzlaf Joe Edward, Mireille Baltzinger, Ite A. Laird-Offringa, Liann Wang, Erica Boni, Tiffany Tsang, Zaneta Nikolovska-Coleska, Andreas Schoenemann, Yuki Abe, Jamie Furneisen, Kenneth T. Lewis, Eileen M. Lafer, John Corbin, Satyen Gautam, Yuguo Feng, Olan Dolezal, Sylviane Hoos, James R. Horn, Bianca Beusink, Jinlin Peng, Otto Pritsch, Kristian H. Schlick, Ryan James Darling, Malgorzata Mikolajczyk, Quincy L. Carter, Jason T. Schuman, Yang Liu, Abdelkrim Khadir, Loïc Martin, Mark Alan Lewis, Ganeshram Krishnamoorthy, Andrew W. Drake, Jason Baardsnes, George Korza, Jesper Pass, Judie Berlier, Melicia Gainey, Nguyen Ly, James R. Partridge, Rosy Calvert, Roberta D'Agata, Rebecca L. Rich, Monica E. Ferreira, Phillip Page, Frank John Podlaski, Jay Duffner, Ruchira Das Gupta, Melanie Wong, Sergei Bibikov, Jiejin Li, Paola Torreri, Bruce A. Andrien, Peter Spies, Christina Boozer, David G. Myszka, University of Utah School of Medicine [Salt Lake City], KaloBios Pharmaceuticals [San Francisco], Schering–Plough Biopharma [Palo Alto], Nomadics [Oklahoma City], Biochemistry and Molecular Biophysics [Pasadena], California Institute of Technology (CALTECH), Department of Biochemistry and Molecular Biophysics, Columbia University [New York], Hartwell Center for Bioinformatics and Biotechnology [Memphis], St. Jude Children’s Research Hospital [Memphis], Universität Zürich [Zürich] = University of Zurich (UZH), Molecular Probes/Invitrogen [Eugene], Institut de génétique et biologie moléculaire et cellulaire (IGBMC), Université Louis Pasteur - Strasbourg I-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), MerckKGaA, Merck & Co. Inc, Abbott Bioresearch Center [Worcester], Institut de biologie cellulaire et moléculaire [Strasbourg], Université de Strasbourg (UNISTRA), Reichert [Depew}, Momenta Pharmaceuticals [Cambridge], XOMA (U.S.), Department of Chemical and Biomolecular Engineering, National University of Singapore, National University of Singapore (NUS), Department of Biology [Ljubljiana], Biotechnical Faculty, University of Ljubljana, Department of Biological Chemistry (Weizmann Institute of Science), Weizmann Institute of Science [Rehovot, Israël], Molecular Biotechnology Core Laboratory [Cleveland], Cleveland Clinic Foundation, ThermoFisher Scientific [Rockford], Biacore/GE Healthcare [Uppsala], AstraZeneca [Hayward], Neurodegeneration Research Department [Harlow], GlaxoSmithKline [Harlow], MedImmune, Biacore/GE Healthcare [Piscataway], Department of Biochemistry [Milwaukee], Medical College of Wisconsin [Milwaukee] (MCW), Alexion Pharmaceuticals [Cheshire], Applied Biosystems [Foster City], diaDexus [South San Francisco], Eli Lilly and Company [Indianapolis], Center for Applied Medical Research [Plamplona] (CIMA), Universidad de Navarra [Pamplona] (UNAV), EMD Lexigen Research Center [Billerica], Department of Cell Biology and Neuroscience [Rome], Istituto Superiore di Sanita [Rome], Département de Biologie structurale et Chimie - Department of Structural Biology and Chemistry, Institut Pasteur [Paris], Department of Chemistry [Atlanta], Georgia State University, University System of Georgia (USG)-University System of Georgia (USG), Pfizer/Rinat Laboratories [South San Francisco], PDLBioPharma [Fremont], Department of Biochemistry [San Antonio], University of Texas Health Science Center at San Antonio [San Antonio], Akubio [Cambridge], ARN : régulations naturelle et artificielle, Université Bordeaux Segalen - Bordeaux 2-Institut Européen de Chimie et de Biologie-Institut National de la Santé et de la Recherche Médicale (INSERM), Wyeth Research [Cambridge], Department of Biochemistry and Molecular Biology [Odense], University of Southern Denmark (SDU), NovoNordisk [Gentofte], Södertörns University College [Huddinge], Department of Biochemistry [Philadelphia], Temple University [Philadelphia], Pennsylvania Commonwealth System of Higher Education (PCSHE)-Pennsylvania Commonwealth System of Higher Education (PCSHE), U.S. Food and Drug Administration (FDA), Department of Biochemical Engineering [London], University College of London [London] (UCL), Merck [Rome], Roche [Palo Alto], University of Twente [Netherlands], Agensys [Santa Monica], Novartis [Emeryville], Massachusetts Institute of Technology (MIT), Northern Illinois University, Institut Pasteur de Montevideo, Réseau International des Instituts Pasteur (RIIP), University of Manchester [Manchester], Department of Physiology [Baltimore], University of Maryland [Baltimore], Complex Carbohydrate Research Center [Athens, GA, USA], University of Georgia [USA], Adnexus Therapeutics [Waltham}, Physiologie de la reproduction et des comportements [Nouzilly] (PRC), Institut National de la Recherche Agronomique (INRA)-Institut Français du Cheval et de l'Equitation [Saumur]-Université de Tours (UT)-Centre National de la Recherche Scientifique (CNRS), ForteBio [Menlo Park], University of Michigan [Ann Arbor], University of Michigan System, Human Genome Sciences [Rockville], Department of Chemical Sciences [Catania], University of Catania [Italy], Montana State University (MSU), GKT School of Biomedical Sciences [London], Organic and Pharmaceutical Chemistry Department [Madrid], Universidad Complutense de Madrid = Complutense University of Madrid [Madrid] (UCM), Bio-Rad Haifa, Institute of Chemistry [Taipei], Academia Sinica, Department of Microbiology and Molecular Genetics, The University of Texas Health Science Center at Houston (UTHealth), Fred Hutchinson Cancer Research Center [Seattle] (FHCRC), Division of Molecular Structure [London], National Institute of Medical Research (UK), Centre for Translational and Chemical Biology, Institute of Structural and Molecular Biology [Edinburgh], University of Edinburgh, Acceleron Pharma [Cambridge], Biotechnology Research Institute [Montreal], National Research Council of Canada (NRC), CSIRO Health Sciences and Nutrition [Parkville], Battelle Biomedical Research Center [Columbus], Attana AB [Stockholm], Corning [USA], School of Life Sciences, Institute for Chemistry and Bioanalytics [Muttenz], University of Applied Sciences Northwestern Switzerland (FHNW), Bio-Rad [Hercules], Monsanto [Chesterfield], Genzyme [Cambridge], Saarland University [Saarbrücken], Institute of Neuroimmunology of SAS [Bratislava], Bristol-Myers Squibb [Princeton], Array Biopharma [Boulder], Service d'Ingénierie Moléculaire pour la Santé (ex SIMOPRO) (SIMoS), Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE)-Université Paris-Saclay-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Department of biochemistry, Masaryk University, Masaryk University [Brno] (MUNI), University of Connecticut Health Center [Farmington], University of Southern California, Los Angeles, GeminX Biotechnologies [Montreal], Hoffman–La Roche [Nutley], IRBM, Pomezia [Rome], Ludwig Institute for Cancer Research, Regeneron Pharmaceuticals [Tarrytown], University of Muenster, Department of Molecular Biology [Salzburg], University of Salzburg, XanTec Bioanalytics [Muenster], Biosciences Division [Los Alamos], Los Alamos National Laboratory (LANL), Lumera [Bothell], Arizona State University [Tempe] (ASU), Dyax [Cambridge], Lombardi Comprehensive Cancer Center, Georgetown University, Department of Biochemistry [Seattle], University of Washington [Seattle], ZymoGenetics, Bio-Rad Canada [Edmonton], Bio-Rad Canada [Toronto], St Jude Children's Research Hospital, University of Ljubljana, Istituto Superiore di Sanità (ISS), Institut Pasteur [Paris] (IP), University of Texas Health Science Center at San Antonio [San Antonio, Tx, USA], University of Twente, Institut National de la Recherche Agronomique (INRA)-Institut Français du Cheval et de l'Equitation [Saumur] (IFCE)-Université de Tours (UT)-Centre National de la Recherche Scientifique (CNRS), University of Southern California (USC), Georgetown University [Washington] (GU), Pasteur Tunis, Institut, Institut National de la Recherche Agronomique (INRA)-Institut Français du Cheval et de l'Equitation [Saumur]-Université de Tours-Centre National de la Recherche Scientifique (CNRS), University of Zürich [Zürich] (UZH), Weizmann Institute of Science, Medical College of Wisconsin [Milwaukee], Centre National de la Recherche Scientifique (CNRS)-Université de Tours-Institut Français du Cheval et de l'Equitation [Saumur]-Institut National de la Recherche Agronomique (INRA), Complutense University of Madrid (UCM), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Paris-Saclay-Direction de Recherche Fondamentale (CEA) (DRF (CEA)), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA), and Masaryk University
- Subjects
Analyte ,Biophysics ,Optical biosensor ,Antibodies, Catalytic ,Biosensing Techniques ,Ligands ,01 natural sciences ,Biochemistry ,Article ,Biacore ,Glutathione transferase ,03 medical and health sciences ,Surface plasmon resonance ,Molecular Biology ,Kinetic rate constant ,030304 developmental biology ,Glutathione Transferase ,0303 health sciences ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Chromatography ,Binding Sites ,Chemistry ,010401 analytical chemistry ,A protein ,Proteins ,Cell Biology ,0104 chemical sciences ,Benchmarking ,Kinetics ,Yield (chemistry) ,Benchmark (computing) ,IR-68798 ,Biosensor ,EWI-16945 ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
International audience; To explore the variability in biosensor studies, 150 participants from 20 countries were given the same protein samples and asked to determine kinetic rate constants for the interaction. We chose a protein system that was amenable to analysis using different biosensor platforms as well as by users of different expertise levels. The two proteins (a 50-kDa Fab and a 60-kDa glutathione S-transferase [GST] antigen) form a relatively high-affinity complex, so participants needed to optimize several experimental parameters, including ligand immobilization and regeneration conditions as well as analyte concentrations and injection/dissociation times. Although most participants collected binding responses that could be fit to yield kinetic parameters, the quality of a few data sets could have been improved by optimizing the assay design. Once these outliers were removed, the average reported affinity across the remaining panel of participants was 620 pM with a standard deviation of 980 pM. These results demonstrate that when this biosensor assay was designed and executed appropriately, the reported rate constants were consistent, and independent of which protein was immobilized and which biosensor was used. (C) 2008 Elsevier Inc. All rights reserved.
- Published
- 2009
- Full Text
- View/download PDF
16. The interaction of cell-penetrating peptides with lipid model systems and subsequent lipid reorganization: thermodynamic and structural characterization
- Author
-
Gérard Chassaing, Emmanuelle Sachon, Isabelle Correia, Isabel D. Alves, Sandrine Sagan, Gordon Tollin, Solange Lavielle, Chen Yu Jiao, Université Pierre et Marie Curie - Paris 6 (UPMC), Service de Biochimie, Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Bicêtre, Biologie intégrative (FRBI), Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Pierre et Marie Curie - Paris 6 (UPMC), Department of Biochemistry and Molecular Biophysics, University of Arizona, Institut de Biologie Intégrative (IFR-BI), Université Pierre et Marie Curie - Paris 6 (UPMC)-Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), and Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Magnetic Resonance Spectroscopy ,Supramolecular chemistry ,Phospholipid ,Peptide ,010402 general chemistry ,01 natural sciences ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Structural Biology ,Drug Discovery ,Amphiphile ,[CHIM]Chemical Sciences ,Molecular Biology ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Pharmacology ,chemistry.chemical_classification ,[PHYS]Physics [physics] ,0303 health sciences ,Calorimetry, Differential Scanning ,Chemistry ,[CHIM.ORGA]Chemical Sciences/Organic chemistry ,Bilayer ,Organic Chemistry ,Cell Membrane ,General Medicine ,Lipids ,0104 chemical sciences ,Membrane ,Drug delivery ,Biophysics ,Cell-penetrating peptide ,Molecular Medicine ,Thermodynamics ,Peptides - Abstract
International audience; Cell-penetrating peptides (CPPs) are cationic peptides that are able to induce cellular uptake and delivery of large and hydrophilic molecules, that otherwise do not cross the plasma membrane of eukaryotic cells. Despite their potential use for gene transfer and drug delivery, the mode of action of CPPs is still mysterious. Nonetheless, the interaction with phospholipid bilayers constitutes the first step in the process. The interaction of two CPPs with distinct charge distribution, penetratin (nonamphipathic) and RL16 (a secondary amphipathic peptide with antimicrobial properties) with lipid membranes was investigated. For this purpose, we employed three independent techniques, comprising (31)P-nuclear magnetic resonance, differential scanning calorimetry (DSC), and plasmon waveguide resonance (PWR) spectroscopy. In view of the cationic nature of the peptides, their interaction and affinity for zwitterionic versus anionic lipids was investigated. Although a strong affinity was observed when negative charged lipids were present, the peptides' thermodynamic behavior on binding to zwitterionic versus anionic lipids and the induced supramolecular structure organization in those lipids was quite different. The study suggests that the amphipathic profile and charge distribution of CPPs strongly influences the perturbation mechanism of the peptide on the bilayer establishing the frontier between a pure CPP and a CPP with antimicrobial properties.
- Published
- 2008
- Full Text
- View/download PDF
17. Joint Composite-Rotation Adiabatic-Sweep Isotope Filtration
- Author
-
Arthur G. Palmer, David Cowburn, Elizabeth R. Valentine, Francesca Massi, Fabien Ferrage, Department of Biochemistry and Molecular Biophysics (DBMB), Columbia University [New York], Biomolécules : synthèse, structure et mode d'action (UMR 8642) (BIOSYMA), Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-École normale supérieure - Paris (ENS Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Pierre et Marie Curie - Paris 6 (UPMC), New York Structural Biology Center (NYSBC), Columbia University [New York]-Wadsworth Center, New York State Department of Health [Albany]-New York State Department of Health [Albany]-New York University [New York] (NYU), NYU System (NYU)-NYU System (NYU)-City University of New York [New York] (CUNY)-Rockefeller University [New York]-Memorial Sloane Kettering Cancer Center [New York]-Icahn School of Medicine at Mount Sinai [New York] (MSSM)- Albert Einstein College of Medicine [New York]-Weill Medical College of Cornell University [New York], École normale supérieure - Paris (ENS-PSL), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS), and Ferrage, Fabien
- Subjects
Magnetic Resonance Spectroscopy ,Analytical chemistry ,[SDV.BBM.BP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biophysics ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Molecular physics ,Article ,[PHYS.PHYS.PHYS-CHEM-PH] Physics [physics]/Physics [physics]/Chemical Physics [physics.chem-ph] ,03 medical and health sciences ,symbols.namesake ,Adiabatic process ,Spectroscopy ,030304 developmental biology ,0303 health sciences ,Carbon Isotopes ,Isotope ,Spins ,Nitrogen Isotopes ,Chemistry ,Proteins ,Reproducibility of Results ,Nuclear magnetic resonance spectroscopy ,0104 chemical sciences ,[CHIM.THEO]Chemical Sciences/Theoretical and/or physical chemistry ,[SDV.BBM.BP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biophysics ,[CHIM.THEO] Chemical Sciences/Theoretical and/or physical chemistry ,Heteronuclear molecule ,Isotopes of carbon ,symbols ,RNA ,[PHYS.PHYS.PHYS-CHEM-PH]Physics [physics]/Physics [physics]/Chemical Physics [physics.chem-ph] ,Hamiltonian (quantum mechanics) ,Two-dimensional nuclear magnetic resonance spectroscopy - Abstract
Joint composite-rotation adiabatic-sweep isotope filter are derived by combining the composite-rotation [A. C. Stuart, K. A. Borzilleri, J. M. Withka, and A. G. Palmer, J. Am. Chem. Soc. 121, 5346–5347 (1999)] and adiabatic-sweep [C. Zwahlen, P. Legault, S. J. F. Vincent, J. Greenblatt, R. Konrat, and L. E. Kay, J. Am. Chem. Soc. 119, 6711–6721 (1997); Ē. Kupče and R. Freeman, J. Magn. Reson. 127, 36–48 (1997)] approaches. The joint isotope filters have improved broadband filtration performance, even for extreme values of the one-bond 1H-13C scalar coupling constants in proteins and RNA molecules. An average Hamiltonian analysis is used to describe evolution of the heteronuclear scalar coupling interaction during the adiabatic sweeps within the isotope filter sequences. The new isotope filter elements permit improved selective detection of NMR resonance signals originating from 1H spins attached to an unlabeled natural abundance component of a complex in which other components are labeled with 13C and 15N isotopes.
- Published
- 2007
18. Suppressors of the egg-laying defective phenotype of sel-12 presenilin mutants implicate the CoREST corepressor complex in LIN-12/Notch signaling in C. elegans
- Author
-
Sophie Jarriault, Iva Greenwald, and Howard Hughes Medical Institute, Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
- Subjects
Embryo, Nonmammalian ,Eggs ,[SDV]Life Sciences [q-bio] ,Polymerase Chain Reaction ,Animals, Genetically Modified ,0302 clinical medicine ,Transgenes ,[SDV.BDD]Life Sciences [q-bio]/Development Biology ,Caenorhabditis elegans ,ComputingMilieux_MISCELLANEOUS ,Genetics ,Regulation of gene expression ,Recombination, Genetic ,0303 health sciences ,Membrane Glycoproteins ,Receptors, Notch ,Helminth Proteins ,Neoplasm Proteins ,DNA-Binding Proteins ,Phenotype ,Lac Operon ,Female ,Signal transduction ,Co-Repressor Proteins ,Research Paper ,Plasmids ,Signal Transduction ,education ,Notch signaling pathway ,Repressor ,Nerve Tissue Proteins ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Biology ,Cell fate determination ,Presenilin ,03 medical and health sciences ,Cornified Envelope Proline-Rich Proteins ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Animals ,Humans ,Caenorhabditis elegans Proteins ,Alleles ,030304 developmental biology ,DNA Primers ,Cell Nucleus ,Membrane Proteins ,Proteins ,biology.organism_classification ,Repressor Proteins ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,Gene Expression Regulation ,Mutation ,RNA ,RNA Splice Sites ,Corepressor ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Presenilin is an essential component of the LIN-12/Notch signaling pathway and also plays a critical role in the genesis of Alzheimer's disease. Previously, a screen for suppressors of the egg-laying defective phenotype caused by partial loss of presenilin activity inCaenorhabditis elegans identified a number of new sprgenes that are potentially involved in the regulation of LIN-12/Notch signaling or presenilin activity. Here we report the molecular identity of two spr genes, spr-1 and spr-5. Our genetic analysis indicates that loss of spr-1 elevateslin-12/Notch gene activity in many different cell fate decisions, suggesting that spr-1 is a negative regulator of LIN-12/Notch signaling. Sequence analysis revealed that spr-1is an ortholog of human CoREST, a known corepressor. SPR-1 is localized to the nucleus and acts in a cell-autonomous manner; furthermore, human CoREST can substitute for SPR-1 in C. elegans. We also show that spr-5 encodes a homolog of p110b, another known member of the CoREST corepressor complex. Our results suggest that the CoREST corepressor complex might be functionally conserved in worms, and we discuss the potential role of SPR-1 and SPR-5 in the repression of transcription of genes involved in, or downstream of, LIN-12/Notch signal transduction.
- Published
- 2002
- Full Text
- View/download PDF
19. The interaction of cell-penetrating peptides with lipid model systems and subsequent lipid reorganization: thermodynamic and structural characterization
- Author
-
I. D. Alves, I. Correira, C.Y. Jiao, E. Sachon, Sandrine SAGAN, S. Lavielle, Tollin, G., Gérard Chassaing, Perron, Nicolas, Université Pierre et Marie Curie - Paris 6 (UPMC), Department of Biochemistry and Molecular Biophysics, USA, and University of Arizona
- Subjects
[CHIM.ORGA]Chemical Sciences/Organic chemistry ,[CHIM.ORGA] Chemical Sciences/Organic chemistry ,ComputingMilieux_MISCELLANEOUS - Abstract
International audience
20. Quantitative and Systematic NMR Measurements of Sequence-Dependent A-T Hoogsteen Dynamics in the DNA Double Helix.
- Author
-
Manghrani A, Rangadurai AK, Szekely O, Liu B, Guseva S, and Al-Hashimi HM
- Abstract
The dynamic properties of DNA depend on the sequence, providing an important source of sequence-specificity in biochemical reactions. However, comprehensively measuring how these dynamics vary with sequence is challenging, especially when they involve lowly populated and short-lived conformational states. Using
1 H CEST supplemented by targeted13 C R NMR experiments, we quantitatively measured Watson-Crick to Hoogsteen dynamics for an A-T base pair in 13 trinucleotide sequence contexts. The Hoogsteen population and exchange rate varied 4-fold and 16-fold, respectively, and were dependent on both the 3'- and 5'-neighbors but only weakly dependent on monovalent ion concentration (25 versus 100 mM NaCl) and pH (6.8 versus 8.0). Flexible TA and CA dinucleotide steps exhibited the highest Hoogsteen populations, and their kinetics rates strongly depended on the 3'-neighbor. In contrast, the stiffer AA and GA steps had the lowest Hoogsteen population, and their kinetics were weakly dependent on the 3'-neighbor. The Hoogsteen lifetime was especially short when G-C neighbors flanked the A-T base pair. Our results uncover a unique conformational basis for sequence-specificity in the DNA double helix and establish the utility of NMR to quantitatively and comprehensively measure sequence-dependent DNA dynamics.1ρ NMR experiments, we quantitatively measured Watson-Crick to Hoogsteen dynamics for an A-T base pair in 13 trinucleotide sequence contexts. The Hoogsteen population and exchange rate varied 4-fold and 16-fold, respectively, and were dependent on both the 3'- and 5'-neighbors but only weakly dependent on monovalent ion concentration (25 versus 100 mM NaCl) and pH (6.8 versus 8.0). Flexible TA and CA dinucleotide steps exhibited the highest Hoogsteen populations, and their kinetics rates strongly depended on the 3'-neighbor. In contrast, the stiffer AA and GA steps had the lowest Hoogsteen population, and their kinetics were weakly dependent on the 3'-neighbor. The Hoogsteen lifetime was especially short when G-C neighbors flanked the A-T base pair. Our results uncover a unique conformational basis for sequence-specificity in the DNA double helix and establish the utility of NMR to quantitatively and comprehensively measure sequence-dependent DNA dynamics.- Published
- 2025
- Full Text
- View/download PDF
21. Season-long microbial dynamics from the cuticle of rice weevil originating at food facilities after dispersal to novel food patches.
- Author
-
Morrison WR 3rd, Ponce MA, Castaldi J, James A, Stoll I, Moreland J, Abshire J, Kim TN, and Gerken AR
- Abstract
Stored-product insects may pose food safety concerns due to their capacity to vector harmful microbes. As climate change progresses, the propensity for vectoring may be affected by temperature. Vectoring capacity may also fluctuate over the season. Thus, we evaluated (i) how the area of microbial growth and morphospecies richness vary over the season from field-collected Sitophilus oryzae that were allowed to disperse onto a novel food patch habitat comprised of agar and (ii) whether temperature in the week preceding collection of S. oryzae affected growth and richness. On a weekly basis during 2022 and 2023, we introduced S. oryzae onto agar, and photographed patches at 3 and 5 d, characterizing growth with ImageJ and visually scoring richness. There was 1.4- to 1.6-fold more microbial growth in patches at 5 d compared to 3 d in both years. The greatest microbial growth consistently occurred from S. oryzae collected during the wheat and maize harvest in grain bins, while morphospecies richness increased progressively over time. We observed an 11-fold and 3-fold increase in the number of morphospecies at the end of the season compared to the beginning in 2022 and 2023. There was 2.1- to 316-fold more microbial growth during the wheat (Jun to Jul) and maize harvest (Sep to Oct) compared to early May. We found a positive exponential relationship between temperature in the field and microbial growth in both years. This study expands our understanding of insect-microbe interactions after harvest and highlights variable periods of risk by food facilities over the season., (Published by Oxford University Press on behalf of Entomological Society of America 2025.)
- Published
- 2025
- Full Text
- View/download PDF
22. Establishing a Top-Down Proteomics Platform on a Time-of-Flight Instrument with Electron-Activated Dissociation.
- Author
-
Searfoss RM, Zahn E, Lin Z, and Garcia BA
- Abstract
Top-down proteomics is the study of intact proteins and their post-translational modifications with mass spectrometry. Historically, this field is more challenging than its bottom-up counterpart because the species are much bigger and have a larger number of possible combinations of sequences and modifications; thus, there is a great need for technological development. With improvements in instrumentation and a multiplicity of fragmentation modes available, top-down proteomics is quickly gaining in popularity with renewed attention on increasing confidence in identification and quantification. Here, we systematically evaluated the Sciex ZenoTOF 7600 system for top-down proteomics, applying standards in the field to validate the platform and further experimenting with its capabilities in electron-activated dissociation and post-translational modification site localization. The instrument demonstrated robustness in standard proteins for platform QC, as aided by zeno trapping. We were also able to apply this to histone post-translational modifications, achieving high sequence coverage that allowed PTM's site localization across protein sequences with optimized EAD fragmentation. We demonstrated the ability to analyze proteins spanning the mass range and included analysis of glycosylated proteins. This is a reference point for future top-down proteomics experiments to be conducted on the ZenoTOF 7600 system.
- Published
- 2025
- Full Text
- View/download PDF
23. Discovery of Human PIM Kinase Inhibitors as a Class of Anthelmintic Drugs to Treat Intestinal Nematode Infections.
- Author
-
Banas V, Elfawal MA, Rosa BA, Mahoney M, Kauffman J, Goetz E, Chen P, Aroian RV, Mitreva M, and Janetka JW
- Subjects
- Animals, Humans, Mice, Structure-Activity Relationship, Trichuris drug effects, Drug Discovery, Intestinal Diseases, Parasitic drug therapy, Ancylostoma drug effects, Trichuriasis drug therapy, Female, Anthelmintics pharmacology, Anthelmintics chemistry, Anthelmintics therapeutic use, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors chemistry, Proto-Oncogene Proteins c-pim-1 antagonists & inhibitors
- Abstract
Soil-transmitted helminth (STH) infections affect one-fourth of the global population and pose a significant threat to human and animal health, with limited treatment options and emerging drug resistance. Trichuris trichiura (whipworm) stands out as a neglected disease, necessitating new drugs to address this unmet medical need. We discovered that several different chemical series of related human Provirus Integration sites for Moloney murine leukemia virus (PIM) family kinase inhibitors possess potent anthelmintic activity by using whole-worm motility assays. Systematic structure-activity relationship (SAR) studies based on the pan -PIM kinase inhibitor CX-6258 were conducted to identify compounds displaying improved in vitro motility inhibition of both adult hookworm ( Ancylostoma ceylanicum ) and adult whipworm ( Trichuris muris ) nematodes. A broad kinase selectivity screen of >450 human kinases confirms PIM1 kinase and others as potential targets for CX-6258 and analogues thereof. In addition, we demonstrated that CX-6258 significantly reduced worm burden and egg counts in the T. muris infection model of mice, establishing it as a new oral small molecule anthelmintic therapeutic.
- Published
- 2025
- Full Text
- View/download PDF
24. TRiC Is a Structural Component of Mammalian Sperm Axonemes.
- Author
-
Brown A, Leung MR, Zeev-Ben-Mordehai T, and Zhang R
- Abstract
The TRiC chaperonin is responsible for folding ~5%-10% of the proteome in eukaryotic cells. Our recent cryo-electron microscopy studies of axonemes from diverse mammalian cell types led to the surprising discovery that a fully assembled TRiC chaperonin is a structural component of mammalian sperm flagella, where it is tethered to the radial spokes of doublet microtubules. In contrast, axoneme-tethered TRiC is not observed in mammalian epithelial cilia, nor in any of the non-mammalian sperm flagella studied to date. In this Perspective, we explore several hypotheses for the potential functions of axoneme-tethered TRiC in mature sperm., (© 2025 The Author(s). Cytoskeleton published by Wiley Periodicals LLC.)
- Published
- 2025
- Full Text
- View/download PDF
25. Metabolic flux analysis to increase oil in seeds.
- Author
-
Mukherjee T, Kambhampati S, Morley SA, Durrett TP, and Allen DK
- Subjects
- Fatty Acids metabolism, Seeds metabolism, Plant Oils metabolism, Metabolic Flux Analysis methods
- Abstract
Ensuring an adequate food supply and enough energy to sustainably support future global populations will require enhanced productivity from plants. Oilseeds can help address these needs; but the fatty acid composition of seed oils is not always optimal, and higher yields are required to meet growing demands. Quantitative approaches including metabolic flux analysis can provide insights on unexpected metabolism (i.e. when metabolism is different than in a textbook) and can be used to guide engineering efforts; however, as metabolism is context specific, it changes with tissue type, local environment, and development. This review describes recent insights from metabolic flux analysis in oilseeds and indicates engineering opportunities based on emerging topics and developing technologies that will aid quantitative understanding of metabolism and enable efforts to produce more oil. We also suggest that investigating the key regulators of fatty acid biosynthesis, such as transcription factors, and exploring metabolic signals like phytohormones in greater depth through flux analysis could open new pathways for advancing genetic engineering and breeding strategies to enhance oil crop production., Competing Interests: Conflict of interest statement. None declared., (© The Author(s) 2024. Published by Oxford University Press on behalf of American Society of Plant Biologists.)
- Published
- 2025
- Full Text
- View/download PDF
26. Structure of a zoonotic H5N1 hemagglutinin reveals a receptor-binding site occupied by an auto-glycan.
- Author
-
Morano NC, Guo Y, Becker JE, Li Z, Yu J, Ho DD, Shapiro L, and Kwong PD
- Subjects
- Humans, Binding Sites, Animals, Antibodies, Neutralizing metabolism, Antibodies, Neutralizing chemistry, Antibodies, Neutralizing immunology, N-Acetylneuraminic Acid metabolism, N-Acetylneuraminic Acid chemistry, Protein Binding, Receptors, Virus metabolism, Receptors, Virus chemistry, Influenza A Virus, H5N1 Subtype metabolism, Influenza A Virus, H5N1 Subtype chemistry, Hemagglutinin Glycoproteins, Influenza Virus metabolism, Hemagglutinin Glycoproteins, Influenza Virus chemistry, Cryoelectron Microscopy, Polysaccharides metabolism, Polysaccharides chemistry, Models, Molecular
- Abstract
Highly pathogenic avian influenza has spilled into many mammals, most notably cows and poultry, with several dozen human breakthrough infections. Zoonotic crossovers, with hemagglutinins mutated to enhance viral ability to use human α2-6-linked sialic acid receptors versus avian α2-3-linked ones, highlight the pandemic risk. To gain insight into these crossovers, we determined the cryoelectron microscopy (cryo-EM) structure of the hemagglutinin from the zoonotic H5N1 A/Texas/37/2024 strain (clade 2.3.4.4b) in complex with a previously reported neutralizing antibody. Surprisingly, we found that the receptor-binding site of this H5N1 hemagglutinin was already occupied by an α2-3-linked sialic acid and that this glycan emanated from asparagine N169 of a neighboring protomer on hemagglutinin itself. This structure thus highlights recognition by influenza hemagglutinin of an "auto"-α2-3-linked sialic acid from N169, an N-linked glycan conserved in 95% of H5 strains, and adds "auto-glycan recognition," which may play a role in viral dispersal, to the complexities surrounding H5N1 zoonosis., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2025 Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
27. Triacylglycerol mobilization underpins mitochondrial stress recovery.
- Author
-
Baker ZN, Zhu Y, Guerra RM, Smith AJ, Arra A, Serrano LR, Overmyer KA, Mukherji S, Craig EA, Coon JJ, and Pagliarini DJ
- Subjects
- Humans, Stress, Physiological, Lipase metabolism, Lipase genetics, Animals, Cardiolipins metabolism, Lipid Droplets metabolism, Doxycycline pharmacology, Mitochondria metabolism, Triglycerides metabolism, Saccharomyces cerevisiae metabolism, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins genetics
- Abstract
Mitochondria are central to myriad biochemical processes, and thus even their moderate impairment could have drastic cellular consequences if not rectified. Here, to explore cellular strategies for surmounting mitochondrial stress, we conducted a series of chemical and genetic perturbations to Saccharomyces cerevisiae and analysed the cellular responses using deep multiomic mass spectrometry profiling. We discovered that mobilization of lipid droplet triacylglycerol stores was necessary for strains to mount a successful recovery response. In particular, acyl chains from these stores were liberated by triacylglycerol lipases and used to fuel biosynthesis of the quintessential mitochondrial membrane lipid cardiolipin to support new mitochondrial biogenesis. We demonstrate that a comparable recovery pathway exists in mammalian cells, which fail to recover from doxycycline treatment when lacking the ATGL lipase. Collectively, our work reveals a key component of mitochondrial stress recovery and offers a rich resource for further exploration of the broad cellular responses to mitochondrial dysfunction., Competing Interests: Competing interests: J.J.C. is a consultant for Thermo Fisher Scientific, 908 Devices, and Seer. The remaining authors declare no competing interests., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
28. Mitochondrial CLPB is a pro-survival factor at the onset of granulocytic differentiation of mouse myeloblastic cells.
- Author
-
Wenta T, Wang G, Van Buren T, Zolkiewski M, and Zolkiewska A
- Subjects
- Animals, Mice, Interleukin-3 metabolism, Interleukin-3 genetics, Interleukin-3 pharmacology, Reactive Oxygen Species metabolism, Membrane Potential, Mitochondrial drug effects, Cell Line, Cell Survival drug effects, Neutrophils metabolism, Neutrophils cytology, Granulocyte Colony-Stimulating Factor metabolism, Granulocyte Colony-Stimulating Factor genetics, Granulocyte Colony-Stimulating Factor pharmacology, Endopeptidase Clp metabolism, Endopeptidase Clp genetics, Mice, Knockout, Cell Differentiation, Mitochondria metabolism, Apoptosis genetics
- Abstract
Loss-of-function mutations in the CLPB gene lead to congenital neutropenia due to impaired neutrophil differentiation. CLPB, a member of the AAA+ family of proteins, resides in the intermembrane space of mitochondria. The mechanism by which a loss of CLPB elicits defects in the differentiation program of neutrophil precursor cells is not understood. Here, we used 32D clone 3 (32Dcl3) cells, an interleukin-3 (IL-3)-dependent mouse myeloblastic cell line model, to investigate the effects of CLPB knockout on myeloblast-to-neutrophil differentiation in vitro. We found that CLPB-deficient 32Dcl3 cells showed a decreased mitochondrial membrane potential and increased levels of insoluble HAX1 aggregates in mitochondria, as compared to control cells. Despite those abnormalities, CLPB loss did not affect cell proliferation rates in the presence of IL-3 but it increased apoptosis after IL-3 withdrawal and simultaneous induction of cell differentiation with granulocytic colony stimulating factor (G-CSF). CLPB-deficient cells that survived the stress associated with IL-3 withdrawal/G-CSF treatment expressed the same levels of differentiation markers as control cells. Moreover, we found that increased apoptosis of CLPB-deficient cells is linked to production of reactive oxygen species (ROS). N-acetylcysteine, exogenous free fatty acids, or exogenous citrate protected CLPB-deficient 32Dcl3 cells from apoptosis at the onset of differentiation. The protective effect of citrate was abolished by inhibition of ATP-citrate lyase (ACLY), an enzyme that converts cytosolic citrate into acetyl-CoA, a substrate for protein acetylation. We propose that citrate supplementation may help mitigate the effects of CLPB loss by facilitating ACLY-dependent ROS detoxification in granulocytic precursor cells., Competing Interests: Declarations. Ethics approval and consent to participate: Not applicable. Competing interests: The authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2025
- Full Text
- View/download PDF
29. LEA_4 motifs function alone and in conjunction with synergistic cosolutes to protect a labile enzyme during desiccation.
- Author
-
Nicholson V, Nguyen K, Gollub E, McCoy M, Yu F, Holehouse AS, Sukenik S, and Boothby TC
- Subjects
- Plant Proteins chemistry, Plant Proteins metabolism, Plant Proteins genetics, Desiccation, Amino Acid Motifs, Citrate (si)-Synthase chemistry, Citrate (si)-Synthase metabolism
- Abstract
Organisms from all kingdoms of life depend on Late Embryogenesis Abundant (LEA) proteins to survive desiccation. LEA proteins are divided into broad families distinguished by the presence of family-specific motif sequences. The LEA_4 family, characterized by 11-residue motifs, plays a crucial role in the desiccation tolerance of numerous species. However, the role of these motifs in the function of LEA_4 proteins is unclear, with some studies finding that they recapitulate the function of full-length LEA_4 proteins in vivo, and other studies finding the opposite result. In this study, we characterize the ability of LEA_4 motifs to protect a desiccation-sensitive enzyme, citrate synthase (CS), from loss of function during desiccation. We show here that LEA_4 motifs not only prevent the loss of function of CS during desiccation but also that they can do so more robustly via synergistically interactions with cosolutes. Our analysis further suggests that cosolutes induce synergy with LEA_4 motifs in a manner that correlates with transfer free energy. This research advances our understanding of LEA_4 proteins by demonstrating that during desiccation their motifs can protect specific clients to varying degrees and that their protective capacity is modulated by their chemical environment. Our findings extend beyond the realm of desiccation tolerance, offering insights into the interplay between IDPs and cosolutes. By investigating the function of LEA_4 motifs, we highlight broader strategies for understanding protein stability and function., (© 2025 The Protein Society.)
- Published
- 2025
- Full Text
- View/download PDF
30. Exploring the interplay between enhancer-promoter interactions and transcription.
- Author
-
Kittle RH and Levo M
- Subjects
- Animals, Humans, Transcriptional Activation genetics, Enhancer Elements, Genetic, Promoter Regions, Genetic, Chromatin genetics, Chromatin metabolism, Transcription, Genetic
- Abstract
Enhancers in metazoan genomes are known to activate their target genes across both short and long genomic distances. Recent advances in chromosome conformation capture assays and single-cell imaging have shed light on the underlying chromatin contacts and dynamics. Yet the relationship between 3D physical enhancer-promoter (E-P) interactions and transcriptional activation remains unresolved. In this brief review, we discuss recent studies exploring this relationship across scales: from developmental stages to the minutes surrounding transcriptional activation and from the tissue level to single-allele subcellular dynamics. We discuss how seemingly contradictory observations might be reconciled and contribute to a refined causal relationship between E-P interactions and transcription, with mutual influences., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
31. Forty sites of TRP channel regulation.
- Author
-
Talyzina IA, Nadezhdin KD, and Sobolevsky AI
- Subjects
- Humans, Binding Sites, Animals, Ligands, Protein Binding, Transient Receptor Potential Channels metabolism, Transient Receptor Potential Channels chemistry
- Abstract
Transient receptor potential (TRP) channels are polymodal molecular sensors that integrate chemical, thermal, mechanical and electrical stimuli and convert them into ionic currents that regulate senses of taste, smell, vision, hearing, touch and contribute to perception of temperature and pain. TRP channels are implicated in the pathogenesis of numerous human diseases, including cancers, and represent one of the most ardently pursued drug targets. Recent advances in structural biology, particularly associated with the cryo-EM "resolution revolution", yielded numerous TRP channel structures in complex with ligands that might have therapeutic potential. In this review, we describe the recent progress in TRP channel structural biology, focusing on the description of identified binding sites for small molecules, their relationship to membrane lipids, and interaction of TRP channels with other proteins. The characterized binding sites and interfaces create a diversity of druggable targets and provide a roadmap to aid in the design of new molecules for tuning TRP channel function in disease conditions., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
32. CLIPA protein pairs function as cofactors for prophenoloxidase activation in Anopheles gambiae.
- Author
-
Wang Y, Jin Q, Kanost MR, and Jiang H
- Subjects
- Animals, Anopheles metabolism, Anopheles genetics, Anopheles enzymology, Catechol Oxidase metabolism, Catechol Oxidase genetics, Enzyme Precursors metabolism, Enzyme Precursors genetics, Insect Proteins metabolism, Insect Proteins genetics
- Abstract
Insect prophenoloxidases (proPO) are activated during immune responses by a proPO activating protease (PAP) in the presence of a high molecular weight cofactor assembled from serine protease homologs (SPH) that lack proteolytic activity. PAPs and the SPHs have a similar architecture, with an amino-terminal clip domain and a carboxyl-terminal protease domain. The SPHs belong to CLIPA subfamily of SP-related proteins. In Manduca sexta, a well characterized biochemical model system for insect immunity, the functional SPH cofactor contains one molecule each from two SPH subfamilies, SPH-I and SPH-II. In Anopheles gambiae, three SPHI-SPHII pairs (CLIPs A4-A6, A4-A7Δ, and A4-A12) were previously reported as cofactors for CLIPB9-mediated activation of proPO2 and proPO7. In this study, we produced recombinant proteins for two splicing variants of CLIPA7, proCLIPA7s (s for short), proCLIPA7f (f for full-length) and proCLIPA14. We cleaved each along with proCLIPA4 using M. sexta PAP3 and found that the CLIPA pairs A4-A7s and A4-A14 are better than A4-A7f in generating highly active PO2 or PO7. CLIPA7f and CLIPA7s, products of alternative splicing, have different strengths as cofactors in combination with CLIPA4. Because mRNA for CLIPA7f is expressed at a significantly higher level than CLIPA7s, cofactors with the weaker combination A4-A7f may predominate in hemolymph, resulting in a potential dampening effect on proPO activation as a regulatory mechanism for altering the strength of the melanization response. A. gambiae CLIPB10x
a is involved in proPO activation but its role as a PAP was not established using mosquito proPOs. Here we showed that factor Xa-treated proCLIPB10Xa activated proCLIPs A7s, A7f, A14, A4 (poorly), and proPO2. At higher concentrations, CLIPB10xa efficiently activated proPO2 in the absence of a cofactor, but at low concentrations it required a CLIPA cofactor, suggesting that highly active PO2 can be generated at low concentration of CLIPB10 in cooperation with an SPH cofactor in vivo. Using cofactors generated by PAP3, we demonstrated the order of efficacy for proPO2 activation by B10Xa is A4-A6 > A4-A14 or A4-A7s > A4-A7f > A4-A12. This agrees with their relative strengths as cofactors for proPO2 and proPO7 activation by M. sexta PAP3. In summary, we further developed an in vitro assay system to elucidate biochemical details of the complex process of proPO activation in A. gambiae., (Copyright © 2025 Elsevier Ltd. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
33. A roadmap to cysteine specific labeling of membrane proteins for single-molecule photobleaching studies.
- Author
-
Ernst M, Mahoney-Kruszka R, Zelt NB, and Robertson JL
- Subjects
- Humans, Fluorescent Dyes chemistry, Maleimides chemistry, Membrane Proteins chemistry, Membrane Proteins metabolism, Single Molecule Imaging methods, Escherichia coli genetics, Escherichia coli metabolism, Protein Multimerization, Escherichia coli Proteins chemistry, Escherichia coli Proteins metabolism, Escherichia coli Proteins genetics, Cysteine chemistry, Cysteine metabolism, Photobleaching
- Abstract
Single-molecule photobleaching analysis is a useful approach for quantifying reactive membrane protein oligomerization in membranes. It provides a binary readout of a fluorophore attached to a protein subunit at dilute conditions. However, quantification of protein stoichiometry from this data requires information about the subunit labeling yields and whether there is non-specific background labeling. Any increases in subunit-specific labeling improves the ability to determine oligomeric states with confidence. A common strategy for site-specific labeling is by conjugation of a fluorophore bearing a thiol-reactive maleimide group to a substituted cysteine. Yet, cysteine reactivity can be difficult to predict as it depends on many factors such as solvent accessibility and electrostatics from the surrounding protein structure. Here we report a general methodology for screening potential cysteine labeling sites on purified membrane proteins. We present the results of two example systems for which the dimerization reactions in membranes have been characterized: (1) the CLC-ec1 Cl
- /H+ antiporter, an Escherichia coli homologue of voltage-gated chloride ion channels in humans and (2) a mutant form of a member of the family of fluoride channels Fluc from Bordetella pertussis (Fluc-Bpe-N43S). To demonstrate how we identify such sites, we first discuss considerations of residue positions hypothesized to be suitable and then describe the specific steps to rigorously assess site-specific labeling while maintaining functional activity and robust single-molecule fluorescence signals. We find that our initial, well rationalized choices are not strong predictors of success, as rigorous testing of the labeling sites shows that only ≈ 30 % of sites end up being useful for single-molecule photobleaching studies., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
34. A multiplex method for rapidly identifying viral protease inhibitors.
- Author
-
Hong SJ, Resnick SJ, Iketani S, Cha JW, Albert BA, Fazekas CT, Chang CW, Liu H, Dagan S, Abagyan MR, Fajtová P, Culbertson B, Brace B, Reddem ER, Forouhar F, Glickman JF, Balkovec JM, Stockwell BR, Shapiro L, O'Donoghue AJ, Sabo Y, Freundlich JS, Ho DD, and Chavez A
- Subjects
- Humans, Antiviral Agents pharmacology, Viral Proteases metabolism, SARS-CoV-2 drug effects, SARS-CoV-2 enzymology, Drug Discovery methods, Biosensing Techniques methods, Coronavirus 3C Proteases antagonists & inhibitors, Coronavirus 3C Proteases metabolism, Protease Inhibitors pharmacology, High-Throughput Screening Assays methods
- Abstract
With current treatments addressing only a fraction of pathogens and new viral threats constantly evolving, there is a critical need to expand our existing therapeutic arsenal. To speed the rate of discovery and better prepare against future threats, we establish a high-throughput platform capable of screening compounds against 40 diverse viral proteases simultaneously. This multiplex approach is enabled by using cellular biosensors of viral protease activity combined with DNA-barcoding technology, as well as several design innovations that increase assay sensitivity and correct for plate-to-plate variation. Among >100,000 compound-target interactions explored within our initial screen, a series of broad-acting inhibitors against coronavirus proteases were uncovered and validated through orthogonal assays. A medicinal chemistry campaign was performed to improve one of the inhibitor's potency while maintaining its broad activity. This work highlights the power of multiplex screening to efficiently explore chemical space at a fraction of the time and costs of previous approaches., Competing Interests: Disclosure and competing interests statement. AC, SJH, and SJR have submitted a patent related to the design and implementation of multiplex small molecule screens. BRS is an inventor on patents and patent applications involving small molecules, holds equity in and serves as a consultant to Exarta Therapeutics and ProJenX Inc., holds equity in Sonata Therapeutics, and serves as a consultant to Weatherwax Biotechnologies Corporation and Akin Gump Strauss Hauer & Feld LLP. DDH is a co-founder of TaiMed Biologics and RenBio, consultant to WuXi Biologics, Brii Biosciences, Apexigen, and Veru Inc., and board director for Vicarious Surgical., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
35. Mechanisms of uropathogenic E. coli mucosal association in the gastrointestinal tract.
- Author
-
Azimzadeh PN, Birchenough GM, Gualbuerto NC, Pinkner JS, Tamadonfar KO, Beatty W, Hannan TJ, Dodson KW, Ibarra EC, Kim S, Schreiber HL 4th, Janetka JW, Kau AL, Earl AM, Miller MJ, Hansson GC, and Hultgren SJ
- Subjects
- Humans, Animals, Mice, Intestinal Mucosa microbiology, Intestinal Mucosa metabolism, Urinary Tract Infections microbiology, Gastrointestinal Tract microbiology, Gastrointestinal Tract metabolism, Bacterial Adhesion, Mucus metabolism, Colon microbiology, Colon metabolism, Epithelial Cells microbiology, Epithelial Cells metabolism, Uropathogenic Escherichia coli, Fimbriae Proteins metabolism, Fimbriae Proteins genetics, Adhesins, Escherichia coli metabolism, Adhesins, Escherichia coli genetics, Escherichia coli Infections microbiology, Escherichia coli Infections metabolism
- Abstract
Urinary tract infections (UTIs) are highly recurrent and frequently caused by Uropathogenic Escherichia coli (UPEC) strains that can be found in patient intestines. Seeding of the urinary tract from this intestinal reservoir likely contributes to UTI recurrence (rUTI) rates. Thus, understanding the factors that promote UPEC intestinal colonization is of critical importance to designing therapeutics to reduce rUTI incidence. Although E. coli is found in high abundance in large intestine mucus, little is known about how it is able to maintain residence in this continuously secreted hydrogel. We discovered that the FimH adhesin of type 1 pili (T1P) bound throughout the secreted mucus layers of the colon and to epithelial cells in mouse and human samples. Disruption of T1P led to reduced association with colon mucus. Notably, this mutant up-regulated flagellar production and infiltrated the protective inner mucus layer of the colon. This could explain how UPEC resists being washed off by the continuously secreted mucus layers of the colon.
- Published
- 2025
- Full Text
- View/download PDF
36. Overexpression of Drosophila NUAK or Constitutively-Active Formin-Like Promotes the Formation of Aberrant Myofibrils.
- Author
-
Tiwari P, Brooks D, and Geisbrecht ER
- Abstract
Muscle development and maintenance is central to the normal functioning of animals. Muscle tissues exhibit high levels of activity and require the dynamic turnover of proteins. An actomyosin scaffold functions with additional proteins comprising the basic contractile subunit of striated muscle, known as the sarcomere. Drosophila muscles are similar to vertebrate muscles in composition and they share a similar mechanism of development. Drosophila NUAK (NUAK) is the homolog of NUAK1 and NUAK2 in vertebrates. NUAK belongs to the family of AMP-activated protein kinases (AMPKs), a group of proteins with broad and overlapping cellular targets. Here we confirm that NUAK dynamically modulates larval muscle sarcomere size as upregulation of NUAK produces longer sarcomeres, including increased thin filament lengths. Furthermore, NUAK overexpression results in aberrant myofibers above the nuclei plane, upregulation of Formin-like (Frl), and an increase in newly synthesized proteins at sites consistent with actin filament assembly. Expression of constitutively-active Frl also produces aberrant myofibers similar to NUAK overexpression. These results taken together strongly suggest a functional link between NUAK and Frl in myofibril formation in an in vivo setting., (© 2025 Wiley Periodicals LLC.)
- Published
- 2025
- Full Text
- View/download PDF
37. Undocking of an extensive ciliary network induces proteostasis and cell fate switching resulting in severe primary ciliary dyskinesia.
- Author
-
Brody SL, Pan J, Huang T, Xu J, Xu H, Koenitizer JR, Brennan SK, Nanjundappa R, Saba TG, Rumman N, Berical A, Hawkins FJ, Wang X, Zhang R, Mahjoub MR, Horani A, and Dutcher SK
- Subjects
- Humans, Proteomics, Microtubules metabolism, Chlamydomonas reinhardtii metabolism, Kartagener Syndrome metabolism, Kartagener Syndrome pathology, Kartagener Syndrome genetics, Axoneme metabolism, Proteins, Cilia metabolism, Proteostasis
- Abstract
Primary ciliary dyskinesia is a rare monogenic syndrome that is associated with chronic respiratory disease, infertility, and laterality defects. Although more than 50 genes causative of primary ciliary dyskinesia have been identified, variants in the genes encoding coiled-coil domain-containing 39 (CCDC39) and CCDC40 in particular cause severe disease that is not explained by loss of ciliary motility alone. Here, we sought to understand the consequences of these variants on cellular functions beyond impaired motility. We used human cells with pathogenic variants in CCDC39 and CCDC40 , Chlamydomonas reinhardtii genetics, cryo-electron microscopy, and proteomics to define perturbations in ciliary assembly and cilia stability, as well as multiple motility-independent pathways. Analysis of proteomics of cilia from patient cells identified that the absence of the axonemal CCDC39/CCDC40 heterodimer resulted in the loss of a network of more than 90 ciliary structural proteins, including 14 that were defined as ciliary address recognition proteins, which provide docking for the missing structures. The absence of the network impaired microtubule architecture, activated cell quality control pathways, switched multiciliated cell fate to mucus-producing cells and resulted in a defective periciliary barrier. In CCDC39 variant cells, these phenotypes were reversed through expression of a normal CCDC39 transgene. These findings indicate that the CCDC39/CCDC40 heterodimer functions as a scaffold to support the assembly of an extensive network of ciliary proteins, whose loss results in both motility-dependent and motility-independent phenotypes that may explain the severity of disease. Gene therapy might be a potential treatment option to be explored in future studies.
- Published
- 2025
- Full Text
- View/download PDF
38. Molecular basis of vitamin K driven γ-carboxylation at membrane interface.
- Author
-
Cao Q, Ammerman A, Saimi M, Lin Z, Shen G, Chen H, Sun J, Chai M, Liu S, Hsu FF, Krezel AM, Gross ML, Xu J, Garcia BA, Liu B, and Li W
- Abstract
The γ-carboxylation of glutamate residues enables Ca
2+ -mediated membrane assembly of protein complexes that support broad physiological functions including hemostasis, calcium homeostasis, immune response, and endocrine regulation1-4 . Modulating γ-carboxylation level provides prevalent treatments for hemorrhagic and thromboembolic diseases5 . This unique posttranslational modification requires vitamin K hydroquinone (KH2 ) to drive highly demanding reactions6 catalyzed by the membrane-integrated γ-carboxylase (VKGC). To decipher underlying mechanisms, we determined cryo-electron microscopy structures of human VKGC in unbound form, with KH2 and four hemostatic and non-hemostatic proteins possessing propeptides and glutamate-rich domains in different carboxylation states. VKGC recognizes substrate proteins via knob-and-hole interactions with propeptides, thereby bringing tethered glutamate-containing segments for processive carboxylation within a large chamber that provides steric control. Propeptide binding also triggers a global conformational change to signal VKGC activation. Through sequential deprotonation and KH2 epoxidation, VKGC generates free hydroxide ion as an exceptionally strong base required to deprotonate the γ-carbon of glutamate for CO2 addition. The diffusion of this superbase, protected and guided by a sealed hydrophobic tunnel, elegantly resolves the challenge of coupling KH2 epoxidation to γ-carboxylation across the membrane interface. These structural insights and extensive functional experiments advance membrane enzymology and propel the development of novel treatments for γ-carboxylation disorders., (© 2025. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2025
- Full Text
- View/download PDF
39. Structural development of the HIV-1 apex-directed PGT145-PGDM1400 antibody lineage.
- Author
-
Mason RD, Zhang B, Morano NC, Shen CH, McKee K, Heimann A, Du R, Nazzari AF, Hodges S, Kanai T, Lin BC, Louder MK, Doria-Rose NA, Zhou T, Shapiro L, Roederer M, Kwong PD, and Gorman J
- Subjects
- Humans, env Gene Products, Human Immunodeficiency Virus immunology, env Gene Products, Human Immunodeficiency Virus chemistry, Cryoelectron Microscopy, Antibodies, Neutralizing immunology, Antibodies, Neutralizing chemistry, Models, Molecular, HEK293 Cells, HIV-1 immunology, HIV Antibodies immunology, HIV Antibodies chemistry
- Abstract
Broadly neutralizing antibodies (bNAbs) targeting the apex of the HIV-1-envelope (Env) trimer comprise the most potent category of HIV-1 bNAbs and have emerged as promising therapeutics. Here, we investigate the development of the HIV-1 apex-directed PGT145-PGDM1400 antibody lineage and report cryo-EM structures at 3.4 Å resolution of PGDM1400 and of an improved PGT145 variant (PGT145-R100aS), each bound to the BG505 Env trimer. Cross-species-based engineering improves PGT145 IC
80 breadth to near that of PGDM1400. Despite similar breadth and potency, the two antibodies differ in their residue-level interactions with important apex features, including N160 glycans and apex cavity, with residue 100i of PGT145 (sulfated tyrosine) penetrating ∼7 Å farther than residue 100i of PGDM1400 (aspartic acid). While apex-directed bNAbs from other donors use maturation pathways that often converge on analogous residue-level recognition, our results demonstrate that divergent residue-level recognition can occur within the same lineage, thereby enabling improved coverage of escape variants., Competing Interests: Declaration of interests The authors declare no competing interests., (Published by Elsevier Inc.)- Published
- 2025
- Full Text
- View/download PDF
40. Investigation of All Disease-Relevant Lysine Acetylation Sites in α-Synuclein Enabled by Non-canonical Amino Acid Mutagenesis.
- Author
-
Shimogawa M, Li MH, Park GSH, Ramirez J, Lee H, Watson PR, Sharma S, Lin Z, Peng C, Garcia BA, Christianson DW, Rhoades E, Eliezer D, and Petersson EJ
- Abstract
Aggregates of α-synuclein (αS) are hallmarks of synucleinopathies, including Parkinson's Disease (PD) and Multiple System Atrophy (MSA). We have recently shown that αS lysine acetylation in the soluble monomer pool varies between healthy controls, PD, and MSA patients. To study the effects of lysine acetylation at all disease-relevant sites of αS, we first compared production of acetylated αS through either native chemical ligation or non-canonical amino acid (ncAA) mutagenesis. Since yields were comparable, ncAA mutagenesis was deemed superior for scanning many acetylation sites. We expressed and purified 12 disease-relevant variants and studied their binding to membranes as well as their aggregation propensities, and found that acetylation of lysine 12, 43, and 80 had particularly strong effects. To understand the implications for acetylation of monomeric αS found in healthy cells, we performed NMR experiments to study protein conformation and fluorescence correlation spectroscopy experiments to quantify lipid binding. We also investigated the effects of acetylation at lysine 12, 43, and 80 on fibril seeding in neurons. Collectively, our biochemical and cell biological investigations indicated that acetylation of K
80 could inhibit aggregation without conferring negative effects on monomer function in healthy cells. Therefore, we studied the structures of fibrils with K80 acetylation through cryo-electron microscopy to uncover the structural basis for these effects. Finally, we identified inhibition of HDAC8 as a way of potentially increasing acetylation at K80 and other inhibitory sites for therapeutic benefit.- Published
- 2025
- Full Text
- View/download PDF
41. Structural and evolutionary insights into the functioning of glycoprotein hormones and their receptors.
- Author
-
Hendrickson WA and Gong Z
- Abstract
The neuroendocrine system that comprises the glycoprotein hormones (GpHs) and their receptors is essential for reproduction and metabolism. Each GpH hormone is an αβ heterodimer of cystine-knot proteins and its cognate receptor is a G-protein coupled receptor (GPCR) distinguished by a large leucine-rich-repeat (LRR) extracellular domain that binds the hormone and a class A GPCR transmembrane domain that signals through an associating heterotrimeric G protein. Hence, the receptors are called LRR-containing GPCRs-LGRs. The vertebrate GpHs and LGRs have co-evolved from homologs in the earliest metazoan animals, including sponges and comb jellies, but these are absent from unicellular organisms and plants. The two GpH subunits and accompanying LGR receptor of the nematode Caenorhabditis elegans are representative of the invertebrate evolutionary predecessors of human GpH proteins and their receptors, for example follicle-stimulating hormone (FSH) and the FSH receptor (FSHR). Atomic structures of the human GpHs and their receptors, which have been determined by X-ray crystallography and cryogenic electron microscopy (cryo-EM), inform the evolutionary process and provide a mechanistic understanding of the transmission of biochemical signals of hormone binding at the cell surface to the elicitation of second messengers such as cyclic AMP in the cytoplasm. There is compelling biochemical and cellular evidence for the importance of receptor dimers in GpH signaling in cells; yet, all of the human receptors are monomeric as defined beautifully by cryo-EM. Fortunately, the LGR of C. elegans is a stable dimer and its structure, when analyzed in the context of structural information from the human counterparts, predicts a hypothetical model for functionally relevant dimeric associations of the human GpH receptors., (© 2025 American Society of Andrology and European Academy of Andrology.)
- Published
- 2025
- Full Text
- View/download PDF
42. Multimodal hierarchical classification of CITE-seq data delineates immune cell states across lineages and tissues.
- Author
-
Caron DP, Specht WL, Chen D, Wells SB, Szabo PA, Jensen IJ, Farber DL, and Sims PA
- Subjects
- Humans, Cell Lineage genetics, Gene Expression Profiling methods, Sequence Analysis, RNA methods, T-Lymphocyte Subsets immunology, T-Lymphocyte Subsets classification, T-Lymphocyte Subsets metabolism, Single-Cell Analysis methods, Transcriptome
- Abstract
Single-cell RNA sequencing (scRNA-seq) is invaluable for profiling cellular heterogeneity and transcriptional states, but transcriptomic profiles do not always delineate subsets defined by surface proteins. Cellular indexing of transcriptomes and epitopes (CITE-seq) enables simultaneous profiling of single-cell transcriptomes and surface proteomes; however, accurate cell-type annotation requires a classifier that integrates multimodal data. Here, we describe multimodal classifier hierarchy (MMoCHi), a marker-based approach for accurate cell-type classification across multiple single-cell modalities that does not rely on reference atlases. We benchmark MMoCHi using sorted T lymphocyte subsets and annotate a cross-tissue human immune cell dataset. MMoCHi outperforms leading transcriptome-based classifiers and multimodal unsupervised clustering in its ability to identify immune cell subsets that are not readily resolved and to reveal subset markers. MMoCHi is designed for adaptability and can integrate annotation of cell types and developmental states across diverse lineages, samples, or modalities., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
43. DNA polymerase zeta can efficiently replicate structures formed by AT/TA repeat sequences and prevent their deletion.
- Author
-
Das M, Hile SE, Brewster J, Boer JL, Bezalel-Buch R, Guo Q, Yang W, Burgers PM, Eckert KA, and Freudenreich CH
- Subjects
- Humans, Chromosome Fragile Sites genetics, Sequence Deletion, Proliferating Cell Nuclear Antigen metabolism, Proliferating Cell Nuclear Antigen genetics, Nucleotidyltransferases metabolism, Nucleotidyltransferases genetics, Nucleotidyltransferases chemistry, AT Rich Sequence genetics, DNA Polymerase III metabolism, DNA Polymerase III genetics, DNA Polymerase III chemistry, DNA Polymerase II metabolism, DNA Polymerase II genetics, DNA Polymerase II chemistry, Repetitive Sequences, Nucleic Acid genetics, DNA metabolism, DNA genetics, DNA chemistry, Inverted Repeat Sequences, DNA-Directed DNA Polymerase metabolism, DNA-Directed DNA Polymerase genetics, DNA Replication, Saccharomyces cerevisiae Proteins metabolism, Saccharomyces cerevisiae Proteins genetics, Saccharomyces cerevisiae genetics
- Abstract
Long AT repeat tracts form non-B DNA structures that stall DNA replication and cause chromosomal breakage. AT repeats are abundant in human common fragile sites (CFSs), genomic regions that undergo breakage under replication stress. Using an in vivo yeast model system containing AT-rich repetitive elements from human CFS FRA16D, we find that DNA polymerase zeta (Pol ζ) is required to prevent breakage and subsequent deletions at hairpin and cruciform forming (AT/TA)n sequences, with little to no role at an (A/T)28 repeat or a control non-structure forming sequence. DNA polymerase eta is not protective for deletions at AT-rich structures, while DNA polymerase delta is protective, but not in a repeat-specific manner. Using purified replicative holoenzymes in vitro, we show that hairpin structures are most inhibitory to yeast DNA polymerase epsilon, whereas yeast and human Pol ζ efficiently synthesize these regions in a stepwise manner. A requirement for the Rev1 protein and the modifiable lysine 164 of proliferating cell nuclear antigen to prevent deletions at AT/TA repeats suggests a mechanism for Pol ζ recruitment. Our results reveal a novel role for Pol ζ in replicating through AT-rich hairpins and suggest a role for Pol ζ in rescue of stalled replication forks caused by DNA structures., (© The Author(s) 2024. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2025
- Full Text
- View/download PDF
44. Molecular determinants of condensate composition.
- Author
-
Holehouse AS and Alberti S
- Subjects
- Humans, Animals, Signal Transduction, Protein Binding, Biomolecular Condensates metabolism, Biomolecular Condensates chemistry
- Abstract
Cells use membraneless compartments to organize their interiors, and recent research has begun to uncover the molecular principles underlying their assembly. Here, we explore how site-specific and chemically specific interactions shape the properties and functions of condensates. Site-specific recruitment involves precise interactions at specific sites driven by partially or fully structured interfaces. In contrast, chemically specific recruitment is driven by complementary chemical interactions without the requirement for a persistent bound-state structure. We propose that site-specific and chemically specific interactions work together to determine the composition of condensates, facilitate biochemical reactions, and regulate enzymatic activities linked to metabolism, signaling, and gene expression. Characterizing the composition of condensates requires novel experimental and computational tools to identify and manipulate the molecular determinants guiding condensate recruitment. Advancing this research will deepen our understanding of how condensates regulate cellular functions, providing valuable insights into cellular physiology and organization., Competing Interests: Declaration of interests S.A. is an advisor on the scientific advisory board of Dewpoint Therapeutics. A.S.H. is an advisor on the scientific advisory board of Prose Foods., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
45. Enhancer Dynamics and Spatial Organization Drive Anatomically Restricted Cellular States in the Human Spinal Cord.
- Author
-
Kandror EK, Wang A, Carriere M, Peterson A, Liao W, Tjärnberg A, Fung JH, Mahbubani KT, Loper J, Pangburn W, Xu Y, Saeb-Parsy K, Rabadan R, Maniatis T, and Rizvi AH
- Abstract
Here, we report the spatial organization of RNA transcription and associated enhancer dynamics in the human spinal cord at single-cell and single-molecule resolution. We expand traditional multiomic measurements to reveal epigenetically poised and bivalent active transcriptional enhancer states that define cell type specification. Simultaneous detection of chromatin accessibility and histone modifications in spinal cord nuclei reveals previously unobserved cell-type specific cryptic enhancer activity, in which transcriptional activation is uncoupled from chromatin accessibility. Such cryptic enhancers define both stable cell type identity and transitions between cells undergoing differentiation. We also define glial cell gene regulatory networks that reorganize along the rostrocaudal axis, revealing anatomical differences in gene regulation. Finally, we identify the spatial organization of cells into distinct cellular organizations and address the functional significance of this observation in the context of paracrine signaling. We conclude that cellular diversity is best captured through the lens of enhancer state and intercellular interactions that drive transitions in cellular state. This study provides fundamental insights into the cellular organization of the healthy human spinal cord., Competing Interests: DECLARATION OF INTERESTS The authors have no competing interests to declare.
- Published
- 2025
- Full Text
- View/download PDF
46. Proteome-wide association studies for blood lipids and comparison with transcriptome-wide association studies.
- Author
-
Zhang D, Gao B, Feng Q, Manichaikul A, Peloso GM, Tracy RP, Durda P, Taylor KD, Liu Y, Johnson WC, Gabriel S, Gupta N, Smith JD, Aguet F, Ardlie KG, Blackwell TW, Gerszten RE, Rich SS, Rotter JI, Scott LJ, Zhou X, and Lee S
- Subjects
- Humans, Polymorphism, Single Nucleotide, Triglycerides blood, Female, Male, Lipid Metabolism genetics, Gene Expression Profiling, Proteomics, Genome-Wide Association Study, Lipids blood, Proteome metabolism, Proteome genetics, Transcriptome
- Abstract
Blood lipid traits are treatable and heritable risk factors for heart disease, a leading cause of mortality worldwide. Although genome-wide association studies (GWASs) have discovered hundreds of variants associated with lipids in humans, most of the causal mechanisms of lipids remain unknown. To better understand the biological processes underlying lipid metabolism, we investigated the associations of plasma protein levels with total cholesterol (TC), triglycerides (TG), high-density lipoprotein (HDL) cholesterol, and low-density lipoprotein (LDL) cholesterol in blood. We trained protein prediction models based on samples in the Multi-Ethnic Study of Atherosclerosis (MESA) and applied them to conduct proteome-wide association studies (PWASs) for lipids using the Global Lipids Genetics Consortium (GLGC) data. Of the 749 proteins tested, 42 were significantly associated with at least one lipid trait. Furthermore, we performed transcriptome-wide association studies (TWASs) for lipids using 9,714 gene expression prediction models trained on samples from peripheral blood mononuclear cells (PBMCs) in MESA and 49 tissues in the Genotype-Tissue Expression (GTEx) project. We found that although PWASs and TWASs can show different directions of associations in an individual gene, 40 out of 49 tissues showed a positive correlation between PWAS and TWAS signed p values across all the genes, which suggests high-level consistency between proteome-lipid associations and transcriptome-lipid associations., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
47. Biophysical mapping of TREM2-ligand interactions reveals shared surfaces for engagement of multiple Alzheimer's disease ligands.
- Author
-
Greven JA, Wydra JR, Greer RA, Zhi C, Price DA, Svoboda JD, Camitta CLM, Washington M, Leung DW, Song Y, Alexander-Brett J, and Brett TJ
- Subjects
- Humans, Ligands, Protein Binding, Binding Sites, Alzheimer Disease metabolism, Alzheimer Disease genetics, Membrane Glycoproteins metabolism, Membrane Glycoproteins genetics, Receptors, Immunologic metabolism, Receptors, Immunologic genetics
- Abstract
TREM2 is a signaling receptor expressed on microglia that has emerged as an important drug target for Alzheimer's disease and other neurodegenerative diseases. While a number of TREM2 ligands have been identified, little is known regarding the structural details of how they engage. To better understand this, we created a protein library of 28 different TREM2 variants that could be used to map interactions with various ligands using biolayer interferometry. The variants are located in previously identified putative binding surfaces on TREM2 called the hydrophobic site, basic site, and site 2. We found that mutations to the hydrophobic site ablated binding to apoE4 and TDP-43. Competition binding experiments indicated that apoE4 and oAβ42 share overlapping binding sites on TREM2. In contrast, binding to C1q was disrupted most strongly by mutations to the basic site, including R46, with some mutations to the hydrophobic site also attenuating binding, thus suggesting a broader mediation of binding across the two sites. Supporting this, competition experiments indicated that C1q binding could be blocked by both apoE and oAβ42. TREM2 binding to IL-34 was mediated by the basic site at a surface centering on R76. Competition binding experiments validated the unique site for IL-34, showing little to no competition with either oAβ42 or apoE4. However, competition experiments between C1q and IL34 suggest that the ligands compete for binding at the basic site. Altogether, our results suggest that TREM2 utilizes the hydrophobic site (consisting of CDR1, CDR2, and CDR3) as a common site to engage multiple ligands, and uses distinct basic sites to engage others. Our findings imply that pharmaceutical strategies targeting these surfaces might be effective to modulate TREM2 functions., Competing Interests: Declarations. Ethics approval and consent to participate: N/A. Competing interests: The authors report no competing interests., (© 2025. The Author(s).)
- Published
- 2025
- Full Text
- View/download PDF
48. Structure and sequence engineering approaches to improve in vivo expression of nucleic acid-delivered antibodies.
- Author
-
Helble M, Chu J, Flowers K, Trachtman AR, Huynh A, Kim A, Shupin N, Hojecki CE, Gary EN, Solieva S, Parzych EM, Weiner DB, Kulp DW, and Patel A
- Subjects
- Humans, Antibodies, Viral immunology, COVID-19 immunology, COVID-19 virology, Protein Engineering methods, Animals, Mutation, Mice, SARS-CoV-2 immunology, SARS-CoV-2 genetics, Antibodies, Monoclonal genetics
- Abstract
Monoclonal antibodies are an important class of biologics with over 160 Food and Drug Administration/European Union-approved drugs. A significant bottleneck to global accessibility of recombinant monoclonal antibodies stems from complexities related to their production, storage, and distribution. Recently, gene-encoded approaches such as mRNA, DNA, or viral delivery have gained popularity, but ensuring biologically relevant levels of antibody expression in the host remains a critical issue. Using a synthetic DNA platform, we investigated the role of antibody structure and sequence toward in vivo expression. SARS-CoV-2 antibody 2196 was recently engineered as a DNA-encoded monoclonal antibody (DMAb-2196). Utilizing an immunoglobulin heavy and light chain "chain-swap" methodology, we interrogated features of DMAb-2196 that can modulate in vivo expression through rational design and structural modeling. Comparing these results to natural variation of antibody sequences resulted in development of an antibody frequency score that aids in the prediction of expression-improving mutations by leveraging antibody repertoire datasets. We demonstrate that a single amino acid mutation identified through this score increases in vivo expression up to 2-fold and that combinations of mutations can also enhance expression. This analysis has led to a generalized pipeline that can unlock the potential for in vivo delivery of therapeutic antibodies across many indications., Competing Interests: Declaration of interests The authors have multiple pending patents in the area of DMAb technology. D.B.W. has received grant funding, participates in industry collaborations, has received speaking honoraria, and has received fees for consulting, including serving on scientific review committees and board series. Remuneration received by D.B.W. includes direct payments and stock or stock options. D.B.W. also discloses the following paid associations with commercial partners: GeneOne (consultant), Geneos (advisory board), AstraZeneca (advisory board, speaker), Inovio (BOD, SRA, Stock), Sanofi (advisory board), and BBI (advisory board)., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
49. A propofol binding site in the voltage sensor domain mediates inhibition of HCN1 channel activity.
- Author
-
Burtscher V, Wang L, Cowgill J, Chen ZW, Edge C, Smith E, Chang Y, Delemotte L, Evers AS, and Chanda B
- Subjects
- Binding Sites, Humans, Potassium Channels metabolism, Potassium Channels chemistry, Potassium Channels genetics, Ion Channel Gating drug effects, Protein Binding, Protein Domains, Mutation, Animals, Propofol pharmacology, Propofol metabolism, Propofol chemistry, Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels metabolism, Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels chemistry, Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels genetics, Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels antagonists & inhibitors, Molecular Dynamics Simulation
- Abstract
Hyperpolarization-activated and cyclic nucleotide-gated (HCN) ion channels are members of the cyclic nucleotide-binding family and are crucial for regulating cellular automaticity in many excitable cells. HCN channel activation contributes to pain perception, and propofol, a widely used anesthetic, acts as an analgesic by inhibiting the voltage-dependent activity of HCN channels. However, the molecular determinants of propofol action on HCN channels remain unknown. Here, we use a propofol-analog photoaffinity labeling reagent to identify propofol binding sites in the human HCN1 isoform. Mass spectrometry analyses combined with molecular dynamics simulations show that a binding pocket is formed by extracellularly facing residues in the S3 and S4 transmembrane segments in the resting voltage-sensor conformation. Mutations of residues within the putative binding pocket mitigate or eliminate voltage-dependent modulation of HCN1 currents by propofol. Together, these findings reveal a conformation-specific propofol binding site that underlies voltage-dependent inhibition of HCN currents and provides a framework for identifying highly specific modulators of HCN channel gating.
- Published
- 2025
- Full Text
- View/download PDF
50. Structure-function analyses of human TRPV6 ancestral and derived haplotypes.
- Author
-
Neuberger A, Shalygin A, Trofimov YA, Veretenenko II, Nadezhdin KD, Krylov NA, Gudermann T, Efremov RG, Chubanov V, and Sobolevsky AI
- Subjects
- Humans, Calmodulin metabolism, Calmodulin genetics, Calmodulin chemistry, Calcium Channels genetics, Calcium Channels metabolism, Calcium Channels chemistry, Structure-Activity Relationship, HEK293 Cells, Protein Binding, Binding Sites, Polymorphism, Single Nucleotide, TRPV Cation Channels genetics, TRPV Cation Channels chemistry, TRPV Cation Channels metabolism, Haplotypes, Cryoelectron Microscopy, Models, Molecular, Calcium metabolism
- Abstract
TRPV6 is a Ca
2+ selective channel that mediates calcium uptake in the gut and contributes to the development and progression of human cancers. TRPV6 is represented by the ancestral and derived haplotypes that differ by three non-synonymous polymorphisms, located in the N-terminal ankyrin repeat domain (C157R), S1-S2 extracellular loop (M378V), and C-terminus (M681T). The ancestral and derived haplotypes were proposed to serve as genomic factors causing a different outcome for cancer patients of African ancestry. We solved cryoelectron microscopy (cryo-EM) structures of ancestral and derived TRPV6 in the open and calmodulin (CaM)-bound inactivated states. Neither state shows substantial structural differences caused by the non-synonymous polymorphisms. Functional properties assessed by electrophysiological recordings and Ca2+ uptake measurements, and water and ion permeation evaluated by molecular modeling also appear similar between the haplotypes. Therefore, ancestral and derived TRPV6 have similar structure and function, implying that other factors are responsible for the differences in susceptibility to cancer., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.