Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(52): e2313009120, 2023 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-38109533

RESUMO

Genetic medicines have the potential to treat various diseases; however, certain ailments including inflammatory diseases and cancer would benefit from control over extracellular localization of therapeutic proteins. A critical gap therefore remains the need to develop and incorporate methodologies that allow for posttranslational control over expression dynamics, localization, and stability of nucleic acid-generated protein therapeutics. To address this, we explored how the body's endogenous machinery controls protein localization through signal peptides (SPs), including how these motifs could be incorporated modularly into therapeutics. SPs serve as a virtual zip code for mRNA transcripts that direct the cell where to send completed proteins within the cell and the body. Utilizing this signaling biology, we incorporated secretory SP sequences upstream of mRNA transcripts coding for reporter, natural, and therapeutic proteins to induce secretion of the proteins into systemic circulation. SP sequences generated secretion of various engineered proteins into the bloodstream following intravenous, intramuscular, and subcutaneous SP mRNA delivery by lipid, polymer, and ionizable phospholipid delivery carriers. SP-engineered etanercept/TNF-α inhibitor proteins demonstrated therapeutic efficacy in an imiquimod-induced psoriasis model by reducing hyperkeratosis and inflammation. An SP-engineered anti-PD-L1 construct mediated mRNA encoded proteins with longer serum half-lives that reduced tumor burden and extended survival in MC38 and B16F10 cancer models. The modular nature of SP platform should enable intracellular and extracellular localization control of various functional proteins for diverse therapeutic applications.


Assuntos
Dermatite , Melanoma , Psoríase , Humanos , Animais , Melanoma/tratamento farmacológico , Melanoma/genética , Psoríase/tratamento farmacológico , Psoríase/genética , Inflamação/patologia , Sinais Direcionadores de Proteínas , RNA Mensageiro/genética , Modelos Animais de Doenças
2.
J Virol ; 96(8): e0003122, 2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35343807

RESUMO

Human cytomegalovirus (HCMV) tropism for epithelial cells is determined by the pentameric glycoprotein complex found on the viral envelope. Laboratory-adapted strains, such as AD169, typically develop loss-of-function mutations for the pentamer, thus losing the ability to efficiently initiate lytic replication in epithelial cells. Using our human salivary gland-derived epithelial (hSGE) cell model, we observed that 3 chemically distinct histone deacetylase (HDAC) inhibitors can rescue infection in hSGE cells using pentamer-null strains of HCMV. Additionally, infection in ARPE-19 epithelial cells was rescued in a similar manner. We isolated nuclei from AD169-infected cells, quantified viral genomes by quantitative PCR (qPCR), and discovered that while HDAC inhibitors increased immediate early (IE) gene expression, they did not increase the amount of viral DNA in the nucleus. Using immunofluorescence microscopy, we observed that pentamer-null strains showed punctate patterning of pp71 in proximity to the nucleus of infected cells, while pp71 was localized to the nucleus after infection with pentamer-containing strains. Upon treatment with HDAC inhibitors, these punctae remained perinuclear, while more cells displayed entry into the lytic cycle, noted by increased IE-positive nuclei. Taken together, our data indicate that HCMV pentamer-null viruses are able to infect epithelial cells (albeit less efficiently than pentamer-positive viruses) and traffic to the nucleus but fail to initiate lytic gene expression once there. These studies reveal a novel postentry function of the pentamer in addition to the recognized role of pentamer in mediating entry. IMPORTANCE Human cytomegalovirus has a wide cellular tropism, which is driven by one of its glycoprotein complexes, the pentamer. Laboratory-adapted strains continuously passaged on fibroblasts readily lose pentamer function and thus lose their ability to infect diverse cell types such as epithelial cells. Pentamer has been attributed an entry function during infection, but mechanistic details as to how this is achieved have not been definitely demonstrated. In this study, we investigate how pharmacological rescue of pentamer-null strains during epithelial infection by histone deacetylase inhibitors implicates a novel role for the pentamer downstream of entry. This work expands on potential functions of the pentamer, will drive future studies to understand mechanistically how it affects tropism, and provides a new target for future therapeutics.


Assuntos
Citomegalovirus , Células Epiteliais , Inibidores de Histona Desacetilases , Proteínas do Envelope Viral , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/metabolismo , Células Epiteliais/virologia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Humanos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus
3.
Proc Natl Acad Sci U S A ; 117(9): 4921-4930, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32071223

RESUMO

Antibiotic-resistant superbug bacteria represent a global health problem with no imminent solutions. Here we demonstrate that the combination (termed AB569) of acidified nitrite (A-NO2-) and Na2-EDTA (disodium ethylenediaminetetraacetic acid) inhibited all Gram-negative and Gram-positive bacteria tested. AB569 was also efficacious at killing the model organism Pseudomonas aeruginosa in biofilms and in a murine chronic lung infection model. AB569 was not toxic to human cell lines at bactericidal concentrations using a basic viability assay. RNA-Seq analyses upon treatment of P. aeruginosa with AB569 revealed a catastrophic loss of the ability to support core pathways encompassing DNA, RNA, protein, ATP biosynthesis, and iron metabolism. Electrochemical analyses elucidated that AB569 produced more stable SNO proteins, potentially explaining one mechanism of bacterial killing. Our data implicate that AB569 is a safe and effective means to kill pathogenic bacteria, suggesting that simple strategies could be applied with highly advantageous therapeutic/toxicity index ratios to pathogens associated with a myriad of periepithelial infections and related disease scenarios.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Ácido Edético/farmacologia , Nitrito de Sódio/farmacologia , Animais , Antibacterianos/uso terapêutico , Biofilmes/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo , Farmacorresistência Bacteriana/efeitos dos fármacos , Ácido Edético/química , Pneumopatias/tratamento farmacológico , Pneumopatias/microbiologia , Redes e Vias Metabólicas , Camundongos , Nitritos/química , Nitritos/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos
4.
Proc Natl Acad Sci U S A ; 116(5): 1755-1764, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30647114

RESUMO

Human cytomegalovirus (HCMV) is a ubiquitous pathogen that undergoes latency in cells of the hematopoietic compartment, although the mechanisms underlying establishment and maintenance of latency remain elusive. We previously reported that the HCMV-encoded G protein-coupled receptor (GPCR) homolog US28 is required for successful latent infection. We now show that US28 protein (pUS28) provided in trans complements the US28Δ lytic phenotype in myeloid cells, suggesting that sustained US28 expression is necessary for long-term latency. Furthermore, expression of pUS28 at the time of infection represses transcription from the major immediate early promoter (MIEP) within 24 h. However, this repression is only maintained in the presence of continual pUS28 expression provided in trans Our data also reveal that pUS28-mediated signaling attenuates both expression and phosphorylation of cellular fos (c-fos), an AP-1 transcription factor subunit, to repress MIEP-driven transcription. AP-1 binds to the MIEP and promotes lytic replication, and in line with this we find that US28Δ infection results in an increase in AP-1 binding to the MIEP, compared with WT latent infection. Pharmacological inhibition of c-fos represses the MIEP during US28Δ infection to levels similar to those we observe during WT latent infection. Together, our data reveal that US28 is required for both establishment and long-term maintenance of HCMV latency, which is modulated, at least in part, by repressing functional AP-1 binding to the MIEP.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/genética , Receptores de Quimiocinas/genética , Receptores Acoplados a Proteínas G/genética , Proteínas Virais/genética , Latência Viral/genética , Linhagem Celular , Regulação Viral da Expressão Gênica/genética , Células HEK293 , Humanos , Regiões Promotoras Genéticas/genética , Transdução de Sinais/genética , Fator de Transcrição AP-1/genética , Replicação Viral/genética
5.
J Virol ; 93(3)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30404806

RESUMO

Various aspects of human cytomegalovirus (HCMV) pathogenesis, including its ability to replicate in specific cells and tissues and the mechanism(s) of horizontal transmission, are not well understood, predominantly because of the strict species specificity exhibited by HCMV. Murine CMV (MCMV), which contains numerous gene segments highly similar to those of HCMV, has been useful for modeling some aspects of CMV pathogenesis; however, it remains essential to build relevant human cell-based systems to investigate how the HCMV counterparts function. The salivary gland epithelium is a site of persistence for both human and murine cytomegaloviruses, and salivary secretions appear to play an important role in horizontal transmission. Therefore, it is important to understand how HCMV is replicating within the glandular epithelial cells so that it might be possible to therapeutically prevent transmission. In the present study, we describe the development of a salivary epithelial model derived from primary human "salispheres." Initial infection of these primary salivary cells with HCMV occurs in a manner similar to that reported for established epithelial lines, in that gH/gL/UL128/UL130/UL131A (pentamer)-positive strains can infect and replicate, while laboratory-adapted pentamer-null strains do not. However, while HCMV enters the lytic phase and produces virus in salivary epithelial cells, it fails to exhibit robust spread throughout the culture and persists in a low percentage of salivary cells. The present study demonstrates the utility of these primary tissue-derived cells for studying HCMV replication in salivary epithelial cells in vitroIMPORTANCE Human cytomegalovirus (HCMV) infects the majority of the world's population, and although it typically establishes a quiescent infection with little to no disease in most individuals, the virus is responsible for a variety of devastating sequelae in immunocompromised adults and in developing fetuses. Therefore, identifying the viral properties essential for replication, spread, and horizontal transmission is an important area of medical science. Our studies use novel human salivary gland-derived cellular models to investigate the molecular details by which HCMV replicates in salivary epithelial cells and provide insight into the mechanisms by which the virus persists in the salivary epithelium, where it gains access to fluids centrally important for horizontal transmission.


Assuntos
Infecções por Citomegalovirus/virologia , Citomegalovirus/genética , Epitélio/virologia , Fibroblastos/virologia , Glândulas Salivares/virologia , Replicação Viral , Células Cultivadas , Infecções por Citomegalovirus/genética , Humanos , Internalização do Vírus
6.
J Endovasc Ther ; 27(3): 473-480, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32242768

RESUMO

Purpose: To evaluate the performance of peripheral intravascular lithotripsy (IVL) in a real-world setting during endovascular treatment of multilevel calcified peripheral artery disease (PAD). Materials and Methods: The Disrupt PAD III Observational Study (ClinicalTrials.gov identifier NCT02923193) is a prospective, nonrandomized, multicenter, single-arm observational study assessing the acute safety and effectiveness of the Shockwave Peripheral IVL System for the treatment of calcified, stenotic lower limb arteries. Patients were eligible if they had claudication or chronic limb-threatening ischemia and moderate or severe arterial calcification. Between November 2017 and August 2018, 200 patients (mean age 72.5±8.7 years; 148 men) were enrolled across 18 sites and followed through hospital discharge. Results: In the 220 target lesions, IVL was more commonly used in combination with other balloon-based technologies (53.8%) and less often with concomitant atherectomy or stenting (19.8% and 29.9%, respectively). There was a 3.4-mm average acute gain at the end of procedure; the final mean residual stenosis was 23.6%. Angiographic complications were rare, with only 2 type D dissections and a single perforation following drug-coated balloon inflation (unrelated to the IVL procedure). There was no abrupt closure, distal embolization, no reflow, or thrombotic event. Conclusion: Use of peripheral IVL to treat severely calcified, stenotic PAD in a real-world study demonstrated low residual stenosis, high acute gain, and a low rate of complications despite the complexity of disease.


Assuntos
Procedimentos Endovasculares , Claudicação Intermitente/terapia , Isquemia/terapia , Litotripsia , Extremidade Inferior/irrigação sanguínea , Doença Arterial Periférica/terapia , Calcificação Vascular/terapia , Idoso , Idoso de 80 Anos ou mais , Doença Crônica , Constrição Patológica , Procedimentos Endovasculares/efeitos adversos , Procedimentos Endovasculares/instrumentação , Feminino , Humanos , Claudicação Intermitente/diagnóstico por imagem , Claudicação Intermitente/fisiopatologia , Isquemia/diagnóstico por imagem , Isquemia/fisiopatologia , Litotripsia/efeitos adversos , Masculino , Pessoa de Meia-Idade , Doença Arterial Periférica/diagnóstico por imagem , Doença Arterial Periférica/fisiopatologia , Estudos Prospectivos , Fatores de Tempo , Resultado do Tratamento , Estados Unidos , Calcificação Vascular/diagnóstico por imagem , Calcificação Vascular/fisiopatologia
7.
Am J Ind Med ; 63(6): 465-477, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32270550

RESUMO

BACKGROUND: Respirable crystalline silica (RCS) can potentially cause silicosis, lung cancer, and renal failure. The current study estimates the percentages of workers potentially overexposed to concentrations of RCS dust and silicosis proportional mortality rates (PMRs) by industry. METHODS: Occupational Safety and Health Administration compliance inspection sampling data for RCS collected during 1979 to 2015 were used to estimate percentages of workers exposed. The results were used in combination with US Census Bureau estimates to produce industry specific worker population estimates for 2014. Estimates of the numbers and percentages of workers exposed to RCS concentrations at least 1, 2, 5, and 10 times the National Institute for Occupational Safety and Health recommended exposure limit (REL) were calculated by industry using the 2002 North American Industry Classification System. Silicosis PMRs by industry were estimated using National Center for Health Statistics multiple cause of death data. RESULTS: RCS concentrations/workers exposed were highest in the poured concrete foundation and structure contractors; commercial and institutional building construction; and masonry contractors. Approximately 100 000 workers were exposed above the RCS REL, and most (79%) worked in the construction industry. Tile and terrazzo contractors (12%); brick, stone, and related construction merchant wholesalers (10%); masonry contractors (6%) and poured concrete foundation and structure contractors (6%) were the highest percentages of workers potentially overexposed. PMRs were highest for the structural clay product manufacturing and the foundries industries. CONCLUSION: Percentages of workers exposed to RCS varied by industry and in some industries workers are exposed over 10 times the REL. Exposures can be reduced below the REL by implementing the hierarchy of controls.


Assuntos
Poluentes Ocupacionais do Ar/análise , Indústrias/estatística & dados numéricos , Exposição por Inalação/análise , Exposição Ocupacional/análise , Dióxido de Silício/análise , Silicose/mortalidade , Poluentes Ocupacionais do Ar/efeitos adversos , Poeira/análise , Monitoramento Ambiental/estatística & dados numéricos , Humanos , Exposição por Inalação/efeitos adversos , Exposição Ocupacional/efeitos adversos , Silicose/etiologia , Estados Unidos/epidemiologia , United States Occupational Safety and Health Administration
8.
J Gen Virol ; 97(11): 2957-2972, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27638684

RESUMO

Cytomegaloviruses (CMVs) produce chemokines (vCXCLs) that have both sequence and functional homology to host chemokines. Assessment of vCXCL-1's role in CMV infection is limited to in vitro and in silico analysis due to CMVs species specificity. In this study, we used the murine CMV (MCMV) mouse model to evaluate the function of vCXCL-1 in vivo. Recombinant MCMVs expressing chimpanzee CMV vCXCL-1 (vCXCL-1CCMV) or host chemokine, mCXCL1, underwent primary dissemination to the popliteal lymph node, spleen and lung similar to the parental MCMV. However, neither of the recombinants expressing chemokines was recovered from the salivary gland (SG) at any time post-infection although viral DNA was detected. This implies that the virus does not grow in the SG or the overexpressed chemokine induces an immune response that leads to suppressed growth. Pointing to immune suppression of virus replication, recombinant viruses were isolated from the SG following infection of immune-ablated mice [i.e. SCID (severe combined immunodeficiency), NSG (non-obese diabetic SCID gamma) or cyclophosphamide treated]. Depletion of neutrophils or NK cells does not rescue the recovery of chemokine-expressing recombinants in the SG. Surprisingly we found that co-infection of parental virus and chemokine-expressing virus leads to the recovery of the recombinants in the SG. We suggest that parental virus reduces the levels of chemokine expression leading to a decrease in inflammatory monocytes and subsequent SG growth. Therefore, aberrant expression of the chemokines induces cells of the innate and adaptive immune system that curtail the growth and dissemination of the recombinants in the SG.


Assuntos
Quimiocinas CXC/imunologia , Infecções por Citomegalovirus/veterinária , Muromegalovirus/imunologia , Glândulas Salivares/virologia , Proteínas Virais/imunologia , Imunidade Adaptativa , Animais , Quimiocinas CXC/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Camundongos , Camundongos SCID , Muromegalovirus/genética , Pan troglodytes , Glândulas Salivares/imunologia , Proteínas Virais/genética
9.
J Virol ; 88(20): 11811-24, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25100846

RESUMO

Human cytomegalovirus (HCMV) is a pathogen found worldwide and is a serious threat to immunocompromised individuals and developing fetuses. Due to the species specificity of cytomegaloviruses, murine cytomegalovirus (MCMV) has been used as a model for in vivo studies of HCMV pathogenesis. The MCMV genome, like the genomes of other beta- and gammaherpesviruses, encodes G protein-coupled receptors (GPCRs) that modulate host signaling pathways presumably to facilitate viral replication and dissemination. Among these viral receptors, the M33 GPCR carried by MCMV is an activator of CREB, NF-κB, and phospholipase C-ß signaling pathways and has been implicated in aspects of pathogenesis in vivo, including persistence in the salivary glands of BALB/c mice. In this study, we used immunocompetent nonobese diabetic (NOD) and immunocompromised NOD-scid-gamma (NSG) mice to further investigate the salivary gland defect exhibited by M33 deficiency. Interestingly, we demonstrate that virus with an M33 deletion (ΔM33) can replicate in the salivary gland of immunocompromised animals, albeit with a 400-fold growth defect compared with the growth of wild-type virus. Moreover, we determined that M33 does not have a role in cell-associated hematogenous dissemination but is required for viral amplification once the virus reaches the salivary gland. We conclude that the reduced replicative capacity of the ΔM33 virus is due to a specific defect occurring within the localized environment of the salivary gland. Importantly, since the salivary gland represents a site essential for persistence and horizontal transmission, an understanding of the mechanisms of viral replication within this site could lead to the generation of novel therapeutics useful for the prevention of HCMV spread. Importance: Human cytomegalovirus infects the majority of the American people and can reside silently in infected individuals for the duration of their lives. Under a number of circumstances, the virus can reactivate, leading to a variety of diseases in both adults and developing babies, and therefore, identifying the function of viral proteins is essential to understand how the virus spreads and causes disease. We aim to utilize animal models to study the function of an important class of viral proteins termed G protein-coupled receptors with the ultimate goal of developing inhibitors to these proteins that could one day be used to prevent viral spread.


Assuntos
Citomegalovirus/crescimento & desenvolvimento , Receptores Acoplados a Proteínas G/genética , Glândulas Salivares/virologia , Animais , Linhagem Celular , Citomegalovirus/genética , Camundongos , Camundongos Endogâmicos BALB C , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Adv Virus Res ; 118: 1-75, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38461029

RESUMO

G protein coupled receptors (GPCRs) are seven-transmembrane domain proteins that modulate cellular processes in response to external stimuli. These receptors represent the largest family of membrane proteins, and in mammals, their signaling regulates important physiological functions, such as vision, taste, and olfaction. Many organisms, including yeast, slime molds, and viruses encode GPCRs. Cytomegaloviruses (CMVs) are large, betaherpesviruses, that encode viral GPCRs (vGPCRs). Human CMV (HCMV) encodes four vGPCRs, including UL33, UL78, US27, and US28. Each of these vGPCRs, as well as their rodent and primate orthologues, have been investigated for their contributions to viral infection and disease. Herein, we discuss how the CMV vGPCRs function during lytic and latent infection, as well as our understanding of how they impact viral pathogenesis.


Assuntos
Infecções por Citomegalovirus , Receptores Acoplados a Proteínas G , Humanos , Animais , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Citomegalovirus/genética , Infecções por Citomegalovirus/metabolismo , Mamíferos/metabolismo
11.
bioRxiv ; 2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38645179

RESUMO

Human cytomegalovirus (HCMV) infects up to 80% of the world's population. Here, we show that HCMV infection leads to widespread changes in human chromatin accessibility and chromatin looping, with hundreds of thousands of genomic regions affected 48 hours after infection. Integrative analyses reveal HCMV-induced perturbation of Hippo signaling through drastic reduction of TEAD1 transcription factor activity. We confirm extensive concordant loss of TEAD1 binding, active H3K27ac histone marks, and chromatin looping interactions upon infection. Our data position TEAD1 at the top of a hierarchy involving multiple altered important developmental pathways. HCMV infection reduces TEAD1 activity through four distinct mechanisms: closing of TEAD1-bound chromatin, reduction of YAP1 and phosphorylated YAP1 levels, reduction of TEAD1 transcript and protein levels, and alteration of TEAD1 exon-6 usage. Altered TEAD1-based mechanisms are highly enriched at genetic risk loci associated with eye and ear development, providing mechanistic insight into HCMV's established roles in these processes.

12.
J Synchrotron Radiat ; 20(Pt 1): 160-5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23254669

RESUMO

Measurements of the spatial variations in the response of three ionization chamber (IC) designs were tested as a function of chamber bias voltage, incident X-ray flux and fill gas. Two components of spatial variation are seen. When the ionization chambers are near saturation, spatial variations exist that are tied to the chamber geometry. While the response of some chambers is relatively flat, others show significant variation across the IC. These variations appear to be inherent in the response of each IC at saturation. When the chamber is far from saturation, large spatial variations in response are present when N(2) is used as a fill gas, but not when ambient air is used as a fill gas. These appear to be tied to space charge effects.

13.
Virus Res ; 330: 199112, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37040820

RESUMO

Human cytomegalovirus (HCMV) is ubiquitous in the human population, infecting >70% of people during the course of their lifetime. HCMV DNA and proteins have been detected in glioblastoma (GBM) tumor samples, but whether the virus is a driver of the malignant process or serendipitous passenger is not well understood. Traditionally, HCMV functions in a cytolytic fashion by proceeding through the lytic cycle and spreading viral particles to other cells. Our study focuses on understanding the pattern of HCMV infection and spread within GBM cells using an in vitro model. In cells derived from a GBM biopsy (U373), we found that HCMV does not spread throughout the culture and, in fact, virus-positive cells rapidly decline over time. Interestingly, the viability of the infected GBM cells remained high over the time course, and this was accompanied by a rapid decline in the number of viral genomes over the same time course. The implications of this atypical infection pattern and how this may affect GBM progression is discussed.


Assuntos
Infecções por Citomegalovirus , Glioblastoma , Humanos , Citomegalovirus/genética , Genoma Viral
14.
Biochemistry ; 51(20): 4126-37, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22551306

RESUMO

Mammalian cell-surface receptors typically display N- or O-linked glycans added post-translationally. Plant lectins such as phytohemagluttinin (PHA) can activate the T cell receptor (TCR) and other cell-surface receptors by binding to glycans and initiating receptor cross-linking. Pathogenic microorganisms such as Bordetella pertussis also express proteins with lectin-like activities. Similar to plant lectins, pertussis toxin (PTx) can activate the TCR and bind to a variety of glycans. However, whether the lectin-like activity of PTx is responsible for its ability to activate TCR signaling has not been formally proven. Here we examined the ability of PTx and a panel of lectins to activate the TCR or a CD8α/CD3ζ chimeric receptor (termed CD8ζ). We demonstrate that CD8ζ rescues PTx-induced signaling events lacking in TCR null cells. This result indicates that CD8ζ can substitute for TCR and supports the hypothesis that PTxB (functioning as a lectin) stimulates signaling via receptor cross-linking rather than by binding to a specific epitope on the TCR. Moreover, PTx is able to activate signaling by binding either N-linked or O-linked glycan-modified receptors as the TCR displays N-linked glycans while CD8ζ displays O-linked glycans. Finally, studies with a diverse panel of lectins indicate that the signaling activity of the lectins does not always correlate with the biochemical reports of ligand preferences. Comparison of lectin signaling through TCR or CD8ζ allows us to better define the structural and functional properties of lectin-glycan interactions using a biologically based signaling readout.


Assuntos
Complexo CD3/química , Antígenos CD8/química , Toxina Pertussis/química , Lectinas de Plantas/química , Linfócitos T/química , Complexo CD3/genética , Complexo CD3/imunologia , Antígenos CD8/genética , Antígenos CD8/imunologia , Engenharia Genética , Humanos , Células Jurkat , Toxina Pertussis/imunologia , Lectinas de Plantas/imunologia , Receptores de Antígenos de Linfócitos T/química , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia
15.
Stat Med ; 31(1): 80-92, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22086761

RESUMO

Latent class analysis can be applied to the outcomes of expert ratings to select objects or subjects that are regarded as prototypical of a category in an ordinal classification system. During a pilot study, Monte Carlo simulations demonstrated that the probability of correct selection is larger when using latent class analysis than when using methods that rely on agreement statistics. Further improvements in the latent class results can also be achieved by applying affine transformations to latent class estimates of sensitivity and specificity. An application is presented that involves the selection of prototypical radiographs.


Assuntos
Sistemas Inteligentes , Radiografia/estatística & dados numéricos , Filme para Raios X/estatística & dados numéricos , Simulação por Computador , Humanos , Método de Monte Carlo , Projetos Piloto
16.
Proc Natl Acad Sci U S A ; 106(16): 6650-5, 2009 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-19363159

RESUMO

Beta-arrestins are multifunctional adaptors that mediate the desensitization, internalization, and some signaling functions of seven-transmembrane receptors (7TMRs). Agonist-stimulated ubiquitination of beta-arrestin2 mediated by the E3 ubiquitin ligase Mdm2 is critical for rapid beta(2)-adrenergic receptor (beta(2)AR) internalization. We now report the discovery that the deubiquitinating enzyme ubiquitin-specific protease 33 (USP33) binds beta-arrestin2 and leads to the deubiquitination of beta-arrestins. USP33 and Mdm2 function reciprocally and favor respectively the stability or lability of the receptor beta-arrestin complex, thus regulating the longevity and subcellular localization of receptor signalosomes. Receptors such as the beta(2)AR, previously shown to form loose complexes with beta-arrestin ("class A") promote a beta-arrestin conformation conducive for binding to the deubiquitinase, whereas the vasopressin V2R, which forms tight beta-arrestin complexes ("class B"), promotes a distinct beta-arrestin conformation that favors dissociation of the enzyme. Thus, USP33-beta-arrestin interaction is a key regulatory step in 7TMR trafficking and signal transmission from the activated receptors to downstream effectors.


Assuntos
Arrestinas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Ubiquitina Tiolesterase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular , Endossomos/efeitos dos fármacos , Endossomos/enzimologia , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Cinética , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos , Vasopressinas/farmacologia , beta-Arrestinas
17.
Elife ; 112022 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-36073784

RESUMO

The mechanistic details of the tethered agonist mode of activation for the adhesion GPCR ADGRF5/GPR116 have not been completely deciphered. We set out to investigate the physiological importance of autocatalytic cleavage upstream of the agonistic peptide sequence, an event necessary for NTF displacement and subsequent receptor activation. To examine this hypothesis, we characterized tethered agonist-mediated activation of GPR116 in vitro and in vivo. A knock-in mouse expressing a non-cleavable GPR116 mutant phenocopies the pulmonary phenotype of GPR116 knock-out mice, demonstrating that tethered agonist-mediated receptor activation is indispensable for function in vivo. Using site-directed mutagenesis and species-swapping approaches, we identified key conserved amino acids for GPR116 activation in the tethered agonist sequence and in extracellular loops 2/3 (ECL2/3). We further highlight residues in transmembrane 7 (TM7) that mediate stronger signaling in mouse versus human GPR116 and recapitulate these findings in a model supporting tethered agonist:ECL2 interactions for GPR116 activation.


Assuntos
Surfactantes Pulmonares , Aminoácidos , Animais , Humanos , Camundongos , Camundongos Knockout , Peptídeos , Surfactantes Pulmonares/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
18.
J Immunol ; 182(9): 5730-9, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380820

RESUMO

Pertussis toxin (PTx) has been shown to exert a variety of effects on immune cells independent of its ability to ADP-ribosylate G proteins. Of these effects, the binding subunit of PTx (PTxB) has been shown to block signaling via the chemokine receptor CCR5, but the mechanism involved in this process is unknown. Here, we show that PTxB causes desensitization of a related chemokine receptor, CXCR4, and explore the mechanism by which this occurs. CXCR4 is the receptor for the chemokine stromal cell-derived factor 1alpha (SDF-1alpha) and elicits a number of biological effects, including stimulation of T cell migration. PTxB treatment causes a decrease in CXCR4 surface expression, inhibits G protein-associated signaling, and blocks SDF-1alpha-mediated chemotaxis. We show that PTxB mediates these effects by activating the TCR signaling network, as the effects are dependent on TCR and ZAP70 expression. Additionally, the activation of the TCR with anti-CD3 mAb elicits a similar set of effects on CXCR4 activity, supporting the idea that TCR signaling leads to cross-desensitization of CXCR4. The inhibition of CXCR4 by PTxB is rapid and transient; however, the catalytic activity of PTx prevents CXCR4 signaling in the long term. Thus, the effects of PTx holotoxin on CXCR4 signaling can be divided into two phases: short term by the B subunit, and long term by the catalytic subunit. These data suggest that TCR crosstalk with CXCR4 is likely a normal cellular process that leads to cross-desensitization, which is exploited by the B subunit of PTx.


Assuntos
Regulação para Baixo/imunologia , Toxina Pertussis/fisiologia , Receptor Cross-Talk/imunologia , Receptores de Antígenos de Linfócitos T/fisiologia , Receptores CXCR4/antagonistas & inibidores , Transdução de Sinais/imunologia , Animais , Bordetella pertussis/imunologia , Bordetella pertussis/patogenicidade , Células CHO , Domínio Catalítico/imunologia , Células Cultivadas , Cricetinae , Cricetulus , Dessensibilização Imunológica , Humanos , Células Jurkat , Receptores de Antígenos de Linfócitos T/deficiência , Receptores de Antígenos de Linfócitos T/genética , Receptores CXCR4/fisiologia
19.
Methods Mol Biol ; 2244: 159-197, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33555587

RESUMO

All of the cytomegaloviruses discovered to date encode two or more genes with significant homology to G protein-coupled receptors (GPCRs). The functions of these cytomegalovirus GPCRs continue to be actively studied and it is clear that they exhibit numerous interesting functions in vitro and in vivo. In this chapter, we review the various methodologies that can be used to examine biochemical aspects of viral GPCR signaling in vitro, as well as examine the biological activity of these viral GPCRs in vitro and in vivo in virus infected cells using recombinant cytomegaloviruses.


Assuntos
Técnicas de Cultura de Células/métodos , Citomegalovirus/genética , Receptores Acoplados a Proteínas G/genética , Animais , Linhagem Celular/virologia , Citomegalovirus/metabolismo , Humanos , Cultura Primária de Células/métodos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia
20.
J Virol ; 83(19): 10016-27, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19605482

RESUMO

The human cytomegalovirus (HCMV)-encoded G-protein-coupled receptor (GPCR) US28 is a potent activator of a number of signaling pathways in HCMV-infected cells. The intracellular carboxy-terminal domain of US28 contains residues critical for the regulation of US28 signaling in heterologous expression systems; however, the role that this domain plays during HCMV infection remains unknown. For this study, we constructed an HCMV recombinant virus encoding a carboxy-terminal domain truncation mutant of US28, FLAG-US28/1-314, to investigate the role that this domain plays in US28 signaling. We demonstrate that US28/1-314 exhibits a more potent phospholipase C-beta (PLC-beta) signal than does wild-type US28, indicating that the carboxy-terminal domain plays an important role in regulating agonist-independent signaling in infected cells. Moreover, HMCV-infected cells expressing the US28/1-314 mutant exhibit a prolonged calcium signal in response to CCL5, indicating that the US28 carboxy-terminal domain also regulates agonist-dependent signaling. Finally, while the chemokine CX3CL1 behaves as an inverse agonist or inhibitor of constitutive US28 signaling to PLC-beta, we demonstrate that CX3CL1 functions as an agonist with regard to US28-stimulated calcium release. This study is the first to demonstrate that the carboxy terminus of US28 controls US28 signaling in the context of HCMV infection and indicates that chemokines such as CX3CL1 can decrease constitutive US28 signals and yet simultaneously promote nonconstitutive US28 signals.


Assuntos
Citomegalovirus/metabolismo , Fibroblastos/virologia , Receptores de Quimiocinas/metabolismo , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Separação Celular , Quimiocina CX3CL1/metabolismo , Cromossomos Artificiais Bacterianos/metabolismo , Citometria de Fluxo , Humanos , Fosfatos de Inositol/química , Modelos Biológicos , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA