Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(6): 1465-1476.e13, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30122350

RESUMEN

Cell-penetrating peptides (CPPs) are short protein segments that can transport cargos into cells. Although CPPs are widely studied as potential drug delivery tools, their role in normal cell physiology is poorly understood. Early during infection, the L2 capsid protein of human papillomaviruses binds retromer, a cytoplasmic trafficking factor required for delivery of the incoming non-enveloped virus into the retrograde transport pathway. Here, we show that the C terminus of HPV L2 proteins contains a conserved cationic CPP that drives passage of a segment of the L2 protein through the endosomal membrane into the cytoplasm, where it binds retromer, thereby sorting the virus into the retrograde pathway for transport to the trans-Golgi network. These experiments define the cell-autonomous biological role of a CPP in its natural context and reveal how a luminal viral protein engages an essential cytoplasmic entry factor.


Asunto(s)
Proteínas de la Cápside/metabolismo , Péptidos de Penetración Celular/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Secuencia de Aminoácidos , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/genética , Endosomas/metabolismo , Aparato de Golgi/virología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/fisiología , Humanos , Mutagénesis , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/genética , Transporte de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Acoplamiento Viral , Internalización del Virus
2.
PLoS Pathog ; 20(6): e1012289, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38829892

RESUMEN

During entry, human papillomavirus (HPV) traffics from the endosome to the trans Golgi network (TGN) and Golgi and then the nucleus to cause infection. Although dynein is thought to play a role in HPV infection, how this host motor recruits the virus to support infection and which entry step(s) requires dynein are unclear. Here we show that the dynein cargo adaptor BICD2 binds to the HPV L2 capsid protein during entry, recruiting HPV to dynein for transport of the virus along the endosome-TGN/Golgi axis to promote infection. In the absence of BICD2 function, HPV accumulates in the endosome and TGN and infection is inhibited. Cell-based and in vitro binding studies identified a short segment near the C-terminus of L2 that can directly interact with BICD2. Our results reveal the molecular basis by which the dynein motor captures HPV to promote infection and identify this virus as a novel cargo of the BICD2 dynein adaptor.


Asunto(s)
Proteínas de la Cápside , Papillomavirus Humano 16 , Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Humanos , Proteínas de la Cápside/metabolismo , Papillomavirus Humano 16/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , Dineínas/metabolismo , Endosomas/metabolismo , Endosomas/virología , Red trans-Golgi/metabolismo , Red trans-Golgi/virología , Internalización del Virus , Unión Proteica , Células HeLa , Proteínas Asociadas a Microtúbulos/metabolismo , Dineínas Citoplasmáticas/metabolismo
3.
Nat Chem Biol ; 20(6): 751-760, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38480980

RESUMEN

Transmembrane (TM) domains as simple as a single span can perform complex biological functions using entirely lipid-embedded chemical features. Computational design has the potential to generate custom tool molecules directly targeting membrane proteins at their functional TM regions. Thus far, designed TM domain-targeting agents have been limited to mimicking the binding modes and motifs of natural TM interaction partners. Here, we demonstrate the design of de novo TM proteins targeting the erythropoietin receptor (EpoR) TM domain in a custom binding topology competitive with receptor homodimerization. The TM proteins expressed in mammalian cells complex with EpoR and inhibit erythropoietin-induced cell proliferation. In vitro, the synthetic TM domain complex outcompetes EpoR homodimerization. Structural characterization reveals that the complex involves the intended amino acids and agrees with our designed molecular model of antiparallel TM helices at 1:1 stoichiometry. Thus, membrane protein TM regions can now be targeted in custom-designed topologies.


Asunto(s)
Proteínas de la Membrana , Unión Proteica , Receptores de Eritropoyetina , Humanos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/química , Receptores de Eritropoyetina/metabolismo , Receptores de Eritropoyetina/química , Modelos Moleculares , Proliferación Celular/efectos de los fármacos , Receptores de Citocinas/metabolismo , Receptores de Citocinas/química , Secuencia de Aminoácidos , Multimerización de Proteína , Animales , Células HEK293
4.
Proc Natl Acad Sci U S A ; 120(42): e2307721120, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37819982

RESUMEN

The activity of proteins is thought to be invariably determined by their amino acid sequence or composition, but we show that a long segment of a viral protein can support infection independent of its sequence or composition. During virus entry, the papillomavirus L2 capsid protein protrudes through the endosome membrane into the cytoplasm to bind cellular factors such as retromer required for intracellular virus trafficking. Here, we show that an ~110 amino acid segment of L2 is predicted to be disordered and that large deletions in this segment abolish infectivity of HPV16 pseudoviruses by inhibiting cytoplasmic protrusion of L2, association with retromer, and proper virus trafficking. The activity of these mutants can be restored by insertion of protein segments with diverse sequences, compositions, and chemical properties, including scrambled amino acid sequences, a tandem array of a short sequence, and the intrinsically disordered region of an unrelated cellular protein. The infectivity of mutants with small in-frame deletions in this segment directly correlates with the size of the segment. These results indicate that the length of the disordered segment, not its sequence or composition, determines its activity during HPV16 pseudovirus infection. We propose that a minimal length of L2 is required for it to protrude far enough into the cytoplasm to bind cytoplasmic trafficking factors, but the sequence of this segment is largely irrelevant. Thus, protein segments can carry out complex biological functions such as Human papillomavirus pseudovirus infection in a sequence-independent manner. This finding has important implications for protein function and evolution.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Humanos , Proteínas de la Cápside/química , Virus del Papiloma Humano , Internalización del Virus , Células HeLa , Cápside/metabolismo , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/química
5.
J Virol ; 98(1): e0179123, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38168672

RESUMEN

In the United States (US), biosafety and biosecurity oversight of research on viruses is being reappraised. Safety in virology research is paramount and oversight frameworks should be reviewed periodically. Changes should be made with care, however, to avoid impeding science that is essential for rapidly reducing and responding to pandemic threats as well as addressing more common challenges caused by infectious diseases. Decades of research uniquely positioned the US to be able to respond to the COVID-19 crisis with astounding speed, delivering life-saving vaccines within a year of identifying the virus. We should embolden and empower this strength, which is a vital part of protecting the health, economy, and security of US citizens. Herein, we offer our perspectives on priorities for revised rules governing virology research in the US.


Asunto(s)
Investigación Biomédica , Contención de Riesgos Biológicos , Virología , Humanos , COVID-19 , Estados Unidos , Virus , Investigación Biomédica/normas
6.
PLoS Pathog ; 19(9): e1011648, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37703297

RESUMEN

Rab GTPases play key roles in controlling intracellular vesicular transport. GTP-bound Rab proteins support vesicle trafficking. Here, we report that, unlike cellular protein cargos, retromer-mediated delivery of human papillomaviruses (HPV) into the retrograde transport pathway during virus entry is inhibited by Rab9a in its GTP-bound form. Knockdown of Rab9a inhibits HPV entry by modulating the HPV-retromer interaction and impairing retromer-mediated endosome-to-Golgi transport of the incoming virus, resulting in the accumulation of HPV in the endosome. Rab9a is in proximity to HPV as early as 3.5 h post-infection, prior to the Rab7-HPV interaction, and HPV displays increased association with retromer in Rab9a knockdown cells, even in the presence of dominant negative Rab7. Thus, Rab9a can regulate HPV-retromer association independently of Rab7. Surprisingly, excess GTP-Rab9a impairs HPV entry, whereas excess GDP-Rab9a reduces association between L2 and Rab9a and stimulates entry. These findings reveal that HPV and cellular proteins utilize the Rab9a host trafficking machinery in distinct ways during intracellular trafficking.


Asunto(s)
Virus del Papiloma Humano , Infecciones por Papillomavirus , Humanos , Internalización del Virus , Infecciones por Papillomavirus/genética , Endosomas , Guanosina Trifosfato
7.
J Virol ; 97(2): e0008923, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36700640

RESUMEN

Viruses have brought humanity many challenges: respiratory infection, cancer, neurological impairment and immunosuppression to name a few. Virology research over the last 60+ years has responded to reduce this disease burden with vaccines and antivirals. Despite this long history, the COVID-19 pandemic has brought unprecedented attention to the field of virology. Some of this attention is focused on concern about the safe conduct of research with human pathogens. A small but vocal group of individuals has seized upon these concerns - conflating legitimate questions about safely conducting virus-related research with uncertainties over the origins of SARS-CoV-2. The result has fueled public confusion and, in many instances, ill-informed condemnation of virology. With this article, we seek to promote a return to rational discourse. We explain the use of gain-of-function approaches in science, discuss the possible origins of SARS-CoV-2 and outline current regulatory structures that provide oversight for virological research in the United States. By offering our expertise, we - a broad group of working virologists - seek to aid policy makers in navigating these controversial issues. Balanced, evidence-based discourse is essential to addressing public concern while maintaining and expanding much-needed research in virology.


Asunto(s)
Investigación , Virología , Virosis , Humanos , COVID-19/prevención & control , Difusión de la Información , Pandemias/prevención & control , Formulación de Políticas , Investigación/normas , Investigación/tendencias , SARS-CoV-2 , Virología/normas , Virología/tendencias , Virosis/prevención & control , Virosis/virología , Virus
8.
Proc Natl Acad Sci U S A ; 117(11): 6121-6128, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32123072

RESUMEN

Virus replication requires critical interactions between viral proteins and cellular proteins that mediate many aspects of infection, including the transport of viral genomes to the site of replication. In human papillomavirus (HPV) infection, the cellular protein complex known as retromer binds to the L2 capsid protein and sorts incoming virions into the retrograde transport pathway for trafficking to the nucleus. Here, we show that short synthetic peptides containing the HPV16 L2 retromer-binding site and a cell-penetrating sequence enter cells, sequester retromer from the incoming HPV pseudovirus, and inhibit HPV exit from the endosome, resulting in loss of viral components from cells and in a profound, dose-dependent block to infection. The peptide also inhibits cervicovaginal HPV16 pseudovirus infection in a mouse model. These results confirm the retromer-mediated model of retrograde HPV entry and validate intracellular virus trafficking as an antiviral target. More generally, inhibiting virus replication with agents that can enter cells and disrupt essential protein-protein interactions may be applicable in broad outline to many viruses.


Asunto(s)
Proteínas de la Cápside/metabolismo , Péptidos de Penetración Celular/farmacología , Papillomavirus Humano 16/efectos de los fármacos , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/tratamiento farmacológico , Internalización del Virus/efectos de los fármacos , Animales , Péptidos de Penetración Celular/uso terapéutico , Cuello del Útero/virología , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Células HeLa , Papillomavirus Humano 16/fisiología , Humanos , Ratones , Infecciones por Papillomavirus/virología , Unión Proteica/efectos de los fármacos , Mapas de Interacción de Proteínas/efectos de los fármacos , Vagina/virología
9.
Genes Dev ; 29(14): 1552-64, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26220997

RESUMEN

Herpesvirus saimiri (HVS) is an oncogenic γ-herpesvirus that produces microRNAs (miRNAs) by cotranscription of precursor miRNA (pre-miRNA) hairpins immediately downstream from viral small nuclear RNAs (snRNA). The host cell Integrator complex, which recognizes the snRNA 3' end processing signal (3' box), generates the 5' ends of HVS pre-miRNA hairpins. Here, we identify a novel 3' box-like sequence (miRNA 3' box) downstream from HVS pre-miRNAs that is essential for miRNA biogenesis. In vivo knockdown and rescue experiments confirmed that the 3' end processing of HVS pre-miRNAs also depends on Integrator activity. Interaction between Integrator and HVS primary miRNA (pri-miRNA) substrates that contain only the miRNA 3' box was confirmed by coimmunoprecipitation and an in situ proximity ligation assay (PLA) that we developed to localize specific transient RNA-protein interactions inside cells. Surprisingly, in contrast to snRNA 3' end processing, HVS pre-miRNA 3' end processing by Integrator can be uncoupled from transcription, enabling new approaches to study Integrator enzymology.


Asunto(s)
Herpesvirus Saimiriino 2/genética , MicroARNs/metabolismo , Procesamiento de Término de ARN 3'/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Herpesvirus Saimiriino 2/metabolismo , Interacciones Huésped-Patógeno/genética , Humanos , Inmunoprecipitación , MicroARNs/química , MicroARNs/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Procesamiento de Término de ARN 3'/genética , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Nuclear Pequeño/metabolismo , Transcripción Genética
10.
PLoS Pathog ; 16(10): e1008946, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33085724

RESUMEN

During internalization and trafficking, human papillomavirus (HPV) moves from the cell surface to the endosome where the transmembrane protease γ-secretase promotes insertion of the viral L2 capsid protein into the endosome membrane. Protrusion of L2 through the endosome membrane into the cytosol allows the recruitment of cytosolic host factors that target the virus to the Golgi en route for productive infection. How endosome-localized HPV is delivered to γ-secretase, a decisive infection step, is unclear. Here we demonstrate that cytosolic p120 catenin, likely via an unidentified transmembrane protein, interacts with HPV at early time-points during viral internalization and trafficking. In the endosome, p120 is not required for low pH-dependent disassembly of the HPV L1 capsid protein from the incoming virion. Rather, p120 is required for HPV to interact with γ-secretase-an interaction that ensures the virus is transported along a productive route. Our findings clarify an enigmatic HPV infection step and provide critical insights into HPV infection that may lead to new therapeutic strategies against HPV-induced diseases.


Asunto(s)
Alphapapillomavirus/patogenicidad , Cateninas/metabolismo , Infecciones por Papillomavirus/virología , Internalización del Virus , Alphapapillomavirus/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proteínas de la Cápside/metabolismo , Endosomas/metabolismo , Células HeLa/virología , Humanos , Membranas Intracelulares/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/metabolismo , Transporte de Proteínas/fisiología , Virión/metabolismo
12.
Mol Pharm ; 17(2): 461-471, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31855437

RESUMEN

Tumor-targeted drug delivery systems offer not only the advantage of an enhanced therapeutic index, but also the possibility of overcoming the limitations that have largely restricted drug design to small, hydrophobic, "drug-like" molecules. Here, we explore the ability of a tumor-targeted delivery system centered on the use of a pH-low insertion peptide (pHLIP) to directly deliver moderately polar, multi-kDa molecules into tumor cells. A pHLIP is a short, pH-responsive peptide capable of inserting across a cell membrane to form a transmembrane helix at acidic pH. pHLIPs target the acidic tumor microenvironment with high specificity, and a drug attached to the inserting end of a pHLIP can be translocated across the cell membrane during the insertion process. We investigate the ability of wildtype pHLIP to deliver peptide nucleic acid (PNA) cargoes of varying sizes across lipid membranes. We find that pHLIP effectively delivers PNAs up to ∼7 kDa into cells in a pH-dependent manner. In addition, pHLIP retains its tumor-targeting capabilities when linked to cargoes of this size, although the amount delivered is reduced for PNA cargoes greater than ∼6 kDa. As drug-like molecules are traditionally restricted to sizes of ∼500 Da, this constitutes an order-of-magnitude expansion in the size range of deliverable drug candidates.


Asunto(s)
Citoplasma/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Melanoma/tratamiento farmacológico , Proteínas de la Membrana/metabolismo , Ácidos Nucleicos de Péptidos/administración & dosificación , Neoplasias Cutáneas/tratamiento farmacológico , Células A549 , Animales , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Concentración de Iones de Hidrógeno , Membrana Dobles de Lípidos/metabolismo , Melanoma/patología , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida/métodos , Neoplasias Cutáneas/patología , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos
13.
Proc Natl Acad Sci U S A ; 114(25): E4961-E4970, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28584122

RESUMEN

The reversible state of proliferative arrest known as "cellular quiescence" plays an important role in tissue homeostasis and stem cell biology. By analyzing the expression of miRNAs and miRNA-processing factors during quiescence in primary human fibroblasts, we identified a group of miRNAs that are induced during quiescence despite markedly reduced expression of Exportin-5, a protein required for canonical miRNA biogenesis. The biogenesis of these quiescence-induced miRNAs is independent of Exportin-5 and depends instead on Exportin-1. Moreover, these quiescence-induced primary miRNAs (pri-miRNAs) are modified with a 2,2,7-trimethylguanosine (TMG)-cap, which is known to bind Exportin-1, and knockdown of Exportin-1 or trimethylguanosine synthase 1, responsible for (TMG)-capping, inhibits their biogenesis. Surprisingly, in quiescent cells Exportin-1-dependent pri-miR-34a is present in the cytoplasm together with a small isoform of Drosha, implying the existence of a different miRNA processing pathway in these cells. Our findings suggest that during quiescence the canonical miRNA biogenesis pathway is down-regulated and specific miRNAs are generated by an alternative pathway to regulate genes involved in cellular growth arrest.


Asunto(s)
Vías Biosintéticas/genética , Proliferación Celular/genética , Carioferinas/genética , MicroARNs/genética , Receptores Citoplasmáticos y Nucleares/genética , Vías Biosintéticas/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citoplasma/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Guanosina/análogos & derivados , Guanosina/farmacología , Células HEK293 , Células HeLa , Humanos , Proteína Exportina 1
14.
Proc Natl Acad Sci U S A ; 114(35): E7262-E7271, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28808001

RESUMEN

The dimeric 44-residue E5 protein of bovine papillomavirus is the smallest known naturally occurring oncoprotein. This transmembrane protein binds to the transmembrane domain (TMD) of the platelet-derived growth factor ß receptor (PDGFßR), causing dimerization and activation of the receptor. Here, we use Rosetta membrane modeling and all-atom molecular dynamics simulations in a membrane environment to develop a chemically detailed model of the E5 protein/PDGFßR complex. In this model, an active dimer of the PDGFßR TMD is sandwiched between two dimers of the E5 protein. Biochemical experiments showed that the major PDGFßR TMD complex in mouse cells contains two E5 dimers and that binding the PDGFßR TMD to the E5 protein is necessary and sufficient to recruit both E5 dimers into the complex. These results demonstrate how E5 binding induces receptor dimerization and define a molecular mechanism of receptor activation based on specific interactions between TMDs.


Asunto(s)
Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/fisiología , Secuencia de Aminoácidos , Animales , Bovinos , Línea Celular , Transformación Celular Viral , Dimerización , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Conformación Molecular , Papillomaviridae/metabolismo , Infecciones por Papillomavirus , Multimerización de Proteína , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo
15.
J Biol Chem ; 293(23): 8787-8801, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29678881

RESUMEN

C-C motif chemokine receptor 5 (CCR5) is a cell surface-associated, immune-regulatory G protein-coupled receptor (GCPR) with seven transmembrane helices. We previously reported the isolation and initial characterization of short artificial transmembrane protein aptamers, named "traptamers," that specifically down-regulate CCR5 expression and inhibit infection of human T cells by HIV strains that use CCR5 as a co-receptor. Here, we investigated the mechanism of traptamer-mediated CCR5 down-regulation and show that most of the traptamers (designated class 1 traptamers) form a stable complex with CCR5 and target it for lysosome-mediated degradation. The ability of these traptamers to down-regulate CCR5 depended on Lys197 in the fifth transmembrane helix of CCR5. In the absence of traptamers, substitution of Lys197 to an uncharged amino acid increased CCR5 stability, and introduction of a lysine at the homologous position in CCR2b, a related chemokine receptor, decreased CCR2b levels. The prototypic class 2 traptamer BY6M4 also formed a complex with CCR5, but CCR5 down-regulation caused by class 2 traptamers did not depend on the lysosome or on Lys197 These results demonstrate that traptamers use diverse mechanisms to down-regulate CCR5 and identify a specific amino acid that plays a central role in controlling chemokine receptor stability. Further studies of these traptamers are likely to provide new insights into CCR5 metabolism and biology and may suggest new therapeutic approaches to modulate the levels of CCR5 and other GPCRs.


Asunto(s)
Aptámeros de Péptidos/farmacología , Lisosomas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Receptores CCR5/metabolismo , Animales , Línea Celular , VIH/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/metabolismo , Humanos , Lisina/análisis , Lisina/metabolismo , Lisosomas/metabolismo , Ratones , Receptores CCR5/química
17.
Annu Rev Microbiol ; 68: 21-43, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24742054

RESUMEN

Many viruses encode short transmembrane proteins that play vital roles in virus replication or virulence. Because many of these proteins are less than 50 amino acids long and not homologous to cellular proteins, their open reading frames were often overlooked during the initial annotation of viral genomes. Some of these proteins oligomerize in membranes and form ion channels. Other miniproteins bind to cellular transmembrane proteins and modulate their activity, whereas still others have an unknown mechanism of action. Based on the underlying principles of transmembrane miniprotein structure, it is possible to build artificial small transmembrane proteins that modulate a variety of biological processes. These findings suggest that short transmembrane proteins provide a versatile mechanism to regulate a wide range of cellular activities, and we speculate that cells also express many similar proteins that have not yet been discovered.


Asunto(s)
Proteínas Virales/metabolismo , Virosis/virología , Virus/genética , Animales , Humanos , Proteínas Virales/química , Proteínas Virales/genética , Virus/química , Virus/metabolismo
18.
RNA ; 22(11): 1785-1792, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27659050

RESUMEN

The proximity ligation assay (PLA) is an immune staining method that detects protein-protein interactions in fixed cells. We describe here RNA-PLA, a simple adaptation of this technology that allows the detection of specific RNA-protein interactions in fixed cells by using a DNA oligonucleotide that hybridizes to a target RNA in combination with an antibody that recognizes the protein bound to the target RNA. Stable and transient RNA-protein interactions can be readily detected by generation of a fluorescent signal in discrete compartments in intact fixed cells with high specificity. We demonstrate that this approach requires the colocalization of the binding protein and its RNA target in the same cellular compartment, use of an oligonucleotide complementary to the target RNA, and the presence of a binding site for the protein in the target RNA.


Asunto(s)
ARN Nuclear Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Compartimento Celular , Células HEK293 , Células HeLa , Humanos
19.
Proc Natl Acad Sci U S A ; 112(34): E4717-25, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26261320

RESUMEN

We have constructed 26-amino acid transmembrane proteins that specifically transform cells but consist of only two different amino acids. Most proteins are long polymers of amino acids with 20 or more chemically distinct side-chains. The artificial transmembrane proteins reported here are the simplest known proteins with specific biological activity, consisting solely of an initiating methionine followed by specific sequences of leucines and isoleucines, two hydrophobic amino acids that differ only by the position of a methyl group. We designate these proteins containing leucine (L) and isoleucine (I) as LIL proteins. These proteins functionally interact with the transmembrane domain of the platelet-derived growth factor ß-receptor and specifically activate the receptor to transform cells. Complete mutagenesis of these proteins identified individual amino acids required for activity, and a protein consisting solely of leucines, except for a single isoleucine at a particular position, transformed cells. These surprisingly simple proteins define the minimal chemical diversity sufficient to construct proteins with specific biological activity and change our view of what can constitute an active protein in a cellular context.


Asunto(s)
Isoleucina/química , Leucina/química , Proteínas/química , Proteínas/fisiología , Secuencia de Aminoácidos , Datos de Secuencia Molecular
20.
J Gen Virol ; 98(11): 2821-2836, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29058661

RESUMEN

Human papillomaviruses (HPVs) are the most common sexually transmitted viruses and one of the most important infectious causes of cancers worldwide. While prophylactic vaccines are effective against certain strains of HPV, established infections still cause deadly cancers in both men and women. HPV traffics to the nucleus via the retrograde transport pathway, but the mechanism of intracellular transport of non-enveloped viruses such as HPV is incompletely understood. Using an overexpression screen, we identify several genes that control HPV16 entry. We focused on the mechanism by which one of the screen hits, stannin, blocks HPV16 infection. Stannin has not been previously implicated in virus entry. Overexpression of stannin specifically inhibits infection by several HPV types, but not other viruses tested. Stannin is constitutively expressed in human keratinocytes, and its basal levels limit entry by HPV16. Stannin is localized to the endolysosomal compartment and does not affect HPV16 binding to cells, virus uptake, or virus uncoating, but inhibits the entry of HPV into the trans-Golgi network (TGN) and stimulates HPV degradation. We further show that stannin interacts with L1 major capsid protein and impairs the interaction of the L2 minor capsid protein with retromer, which is required for virus trafficking to the TGN. Our findings shed light on a novel cellular protein that interferes with HPV entry and highlight the role of retrograde transport in HPV entry.


Asunto(s)
Proteínas de la Cápside/metabolismo , Papillomavirus Humano 16/fisiología , Queratinocitos/inmunología , Queratinocitos/virología , Neuropéptidos/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Internalización del Virus , Animales , Línea Celular , Papillomavirus Humano 16/inmunología , Humanos , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA