Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 362
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 47(3): 510-523.e4, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28930661

RESUMEN

Within the interleukin 1 (IL-1) cytokine family, IL-1 receptor accessory protein (IL-1RAcP) is the co-receptor for eight receptor-cytokine pairs, including those involving cytokines IL-1ß and IL-33. Unlike IL-1ß, IL-33 does not have a signaling complex that includes both its cognate receptor, ST2, and the shared co-receptor IL-1RAcP, which we now present here. Although the IL-1ß and IL-33 complexes shared structural features and engaged identical molecular surfaces of IL-1RAcP, these cytokines had starkly different strategies for co-receptor engagement and signal activation. Our data suggest that IL-1ß binds to IL-1RI to properly present the cytokine to IL-1RAcP, whereas IL-33 binds to ST2 in order to conformationally constrain the cognate receptor in an IL-1RAcP-receptive state. These findings indicate that members of the IL-1 family of cytokines use distinct molecular mechanisms to signal through their shared co-receptor, and they provide the foundation from which to design new therapies to target IL-33 signaling.


Asunto(s)
Interleucina-1/metabolismo , Receptores de Interleucina-1/metabolismo , Transducción de Señal , Animales , Sitios de Unión , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Interleucina-1/química , Proteína 1 Similar al Receptor de Interleucina-1/química , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/química , Interleucina-33/metabolismo , Ratones , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Mutación , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Receptores de Interleucina-1/química , Receptores de Interleucina-1/genética
2.
J Proteome Res ; 23(5): 1649-1665, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38574199

RESUMEN

Plant-based adhesives, such as those made from wheat, have been prominently used for books and paper-based objects and are also used as conservation adhesives. Starch paste originates from starch granules, whereas flour paste encompasses the entire wheat endosperm proteome, offering strong adhesive properties due to gluten proteins. From a conservation perspective, understanding the precise nature of the adhesive is vital as the longevity, resilience, and reaction to environmental changes can differ substantially between starch- and flour-based pastes. We devised a proteomics method to discern the protein content of these pastes. Protocols involved extracting soluble proteins using 0.5 M NaCl and 30 mM Tris-HCl solutions and then targeting insoluble proteins, such as gliadins and glutenins, with a buffer containing 7 M urea, 2 M thiourea, 4% CHAPS, 40 mM Tris, and 75 mM DTT. Flour paste's proteome is diverse (1942 proteins across 759 groups), contrasting with starch paste's predominant starch-associated protein makeup (218 proteins in 58 groups). Transformation into pastes reduces proteomes' complexity. Testing on historical bookbindings confirmed the use of flour-based glue, which is rich in gluten and serpins. High levels of deamidation were detected, particularly for glutamine residues, which can impact the solubility and stability of the glue over time. The mass spectrometry proteomics data have been deposited to the ProteomeXchange, Consortium (http://proteomecentral.proteomexchange.org) via the MassIVE partner repository with the data set identifier MSV000093372 (ftp://MSV000093372@massive.ucsd.edu).


Asunto(s)
Adhesivos , Harina , Glútenes , Proteoma , Almidón , Triticum , Triticum/química , Harina/análisis , Almidón/química , Proteoma/análisis , Proteoma/química , Adhesivos/química , Glútenes/química , Glútenes/análisis , Proteómica/métodos , Proteínas de Plantas/análisis , Gliadina/química , Gliadina/análisis
3.
J Biol Chem ; 299(12): 105353, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37858677

RESUMEN

The PD-1/PD-L1 checkpoint pathway is important for regulating immune responses and can be targeted by immunomodulatory drugs to treat a variety of immune disorders. However, the precise protein-protein interactions required for the initiation of PD-1/PD-L1 signaling are currently unknown. Previously, we designed a series of first-generation PD-1 targeting peptides based on the native interface region of programmed death ligand 1 (PD-L1) that effectively reduced PD-1/PD-L1 binding. In this work, we further characterized the previously identified lead peptide, MN1.1, to identify key PD-1 binding residues and design an optimized peptide, MN1.4. We show MN1.4 is significantly more stable than MN1.1 in serum and retains the ability to block PD-1/PD-L1 complex formation. We further characterized the immunomodulatory effects of MN1.4 treatment by measuring markers of T cell activation in a co-culture model with ovarian cancer cells and peripheral blood mononuclear cells. We found MN1.4 treatment reduced cytokine secretion and suppressed T cell responses in a similar manner as recombinant PD-L1. Therefore, the PD-L1 interface region used to design MN1.4 appeared sufficient to initiate PD-1 signaling and likely represents the minimum necessary region of PD-L1 required for PD-1 recognition. We propose a peptide agonist for PD-1, such as MN1.4, could have several applications for treating autoimmune disorders caused by PD-1 deficiencies such as type 1 diabetes, inflammatory arthritis, or autoimmune side effects arising from monoclonal antibody-based cancer immunotherapies.


Asunto(s)
Antígeno B7-H1 , Modelos Moleculares , Neoplasias , Transducción de Señal , Humanos , Antígeno B7-H1/química , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Inmunoterapia , Leucocitos Mononucleares/metabolismo , Neoplasias/tratamiento farmacológico , Péptidos/farmacología , Receptor de Muerte Celular Programada 1/agonistas , Receptor de Muerte Celular Programada 1/química , Receptor de Muerte Celular Programada 1/metabolismo , Unión Proteica , Mutación , Estructura Cuaternaria de Proteína , Línea Celular Tumoral , Inmunidad/efectos de los fármacos
4.
Int J Mol Sci ; 23(19)2022 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-36232786

RESUMEN

ApoB-100 is a member of a large lipid transfer protein superfamily and is one of the main apolipoproteins found on low-density lipoprotein (LDL) and very low-density lipoprotein (VLDL) particles. Despite its clinical significance for the development of cardiovascular disease, there is limited information on apoB-100 structure. We have developed a novel method based on the "divide and conquer" algorithm, using PSIPRED software, by dividing apoB-100 into five subunits and 11 domains. Models of each domain were prepared using I-TASSER, DEMO, RoseTTAFold, Phyre2, and MODELLER. Subsequently, we used disuccinimidyl sulfoxide (DSSO), a new mass spectrometry cleavable cross-linker, and the known position of disulfide bonds to experimentally validate each model. We obtained 65 unique DSSO cross-links, of which 87.5% were within a 26 Å threshold in the final model. We also evaluated the positions of cysteine residues involved in the eight known disulfide bonds in apoB-100, and each pair was measured within the expected 5.6 Å constraint. Finally, multiple domains were combined by applying constraints based on detected long-range DSSO cross-links to generate five subunits, which were subsequently merged to achieve an uninterrupted architecture for apoB-100 around a lipoprotein particle. Moreover, the dynamics of apoB-100 during particle size transitions was examined by comparing VLDL and LDL computational models and using experimental cross-linking data. In addition, the proposed model of receptor ligand binding of apoB-100 provides new insights into some of its functions.


Asunto(s)
Apolipoproteínas B , Cisteína , Apolipoproteína B-100 , Apolipoproteínas B/metabolismo , Simulación por Computador , Disulfuros , Ligandos , Lipoproteínas LDL/química , Lipoproteínas VLDL , Modelos Estructurales , Sulfóxidos
5.
J Infect Dis ; 223(10): 1677-1680, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33718952

RESUMEN

A cohort consisting of asymptomatic healthcare workers donated temporal serum samples after infection with severe acute respiratory syndrome coronavirus 2. Analysis shows that all asymptomatic healthcare workers had neutralizing antibodies, that these antibodies persist for ≥60 days, and that anti-spike receptor-binding domain immunoglobulin G levels were correspondingly durable over the same time period.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , COVID-19/inmunología , SARS-CoV-2/inmunología , Enfermedades Asintomáticas , COVID-19/epidemiología , Prueba de Ácido Nucleico para COVID-19 , Estudios de Cohortes , Ensayo de Inmunoadsorción Enzimática , Femenino , Personal de Salud , Humanos , Masculino , Pruebas de Neutralización , Encuestas y Cuestionarios , Factores de Tiempo , Virginia/epidemiología
6.
Retrovirology ; 18(1): 6, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33622348

RESUMEN

BACKGROUND: The Human T-cell Lymphotropic Virus Type-1 (HTLV-1) is a blood-borne pathogen and etiological agent of Adult T-cell Leukemia/Lymphoma (ATLL) and HTLV-1 Associated Myelopathy/Tropical Spastic Paraparesis (HAM/TSP). HTLV-1 has currently infected up to 10 million globally with highly endemic areas in Japan, Africa, the Caribbean and South America. We have previously shown that Extracellular Vesicles (EVs) enhance HTLV-1 transmission by promoting cell-cell contact. RESULTS: Here, we separated EVs into subpopulations using differential ultracentrifugation (DUC) at speeds of 2 k (2000×g), 10 k (10,000×g), and 100 k (100,000×g) from infected cell supernatants. Proteomic analysis revealed that EVs contain the highest viral/host protein abundance in the 2 k subpopulation (2 k > 10 k > 100 k). The 2 k and 10 k populations contained viral proteins (i.e., p19 and Tax), and autophagy proteins (i.e., LC3 and p62) suggesting presence of autophagosomes as well as core histones. Interestingly, the use of 2 k EVs in an angiogenesis assay (mesenchymal stem cells + endothelial cells) caused deterioration of vascular-like-tubules. Cells commonly associated with the neurovascular unit (i.e., astrocytes, neurons, and macrophages) in the blood-brain barrier (BBB) showed that HTLV-1 EVs may induce expression of cytokines involved in migration (i.e., IL-8; 100 k > 2 k > 10 k) from astrocytes and monocyte-derived macrophages (i.e., IL-8; 2 k > 10 k). Finally, we found that EVs were able to promote cell-cell contact and viral transmission in monocytic cell-derived dendritic cell. The EVs from both 2 k and 10 k increased HTLV-1 spread in a humanized mouse model, as evidenced by an increase in proviral DNA and RNA in the Blood, Lymph Node, and Spleen. CONCLUSIONS: Altogether, these data suggest that various EV subpopulations induce cytokine expression, tissue damage, and viral spread.


Asunto(s)
Células Endoteliales/virología , Vesículas Extracelulares/virología , Virus Linfotrópico T Tipo 1 Humano/fisiología , Animales , Comunicación Celular , Citocinas/análisis , Citocinas/genética , Citocinas/inmunología , Vesículas Extracelulares/inmunología , Vesículas Extracelulares/fisiología , Femenino , Infecciones por HTLV-I/virología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Proteómica , Células THP-1 , Células U937
7.
Br J Cancer ; 124(9): 1556-1565, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33658639

RESUMEN

BACKGROUND: Circulating tumour DNA (ctDNA) is known as a tumour-specific personalised biomarker, but the mutation-selection criteria from heterogeneous tumours remain a challenge. METHODS: We conducted multiregional sequencing of 42 specimens from 14 colorectal tumours of 12 patients, including two double-cancer cases, to identify mutational heterogeneity to develop personalised ctDNA assays using 175 plasma samples. RESULTS: "Founder" mutations, defined as a mutation that is present in all regions of the tumour in a binary manner (i.e., present or absent), were identified in 12/14 tumours. In contrast, "truncal" mutations, which are the first mutation that occurs prior to the divergence of branches in the phylogenetic tree using variant allele frequency (VAF) as continuous variables, were identified in 12/14 tumours. Two tumours without founder and truncal mutations were hypermutators. Most founder and truncal mutations exhibited higher VAFs than "non-founder" and "branch" mutations, resulting in a high chance to be detected in ctDNA. In post-operative long-term observation for 10/12 patients, early relapse prediction, treatment efficacy and non-relapse corroboration were achievable from frequent ctDNA monitoring. CONCLUSIONS: A single biopsy is sufficient to develop custom dPCR probes for monitoring tumour burden in most CRC patients. However, it may not be effective for those with hypermutated tumours.


Asunto(s)
Biomarcadores de Tumor/genética , ADN Tumoral Circulante/genética , Neoplasias Colorrectales/genética , Cirugía Colorrectal/mortalidad , Mutación , Recurrencia Local de Neoplasia/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/cirugía , Estudios de Seguimiento , Humanos , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/cirugía , Pronóstico , Tasa de Supervivencia , Carga Tumoral
8.
Expert Rev Proteomics ; 18(10): 845-861, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34607525

RESUMEN

INTRODUCTION: Laser Capture Microdissection (LCM) uses a laser to isolate, or capture, specific cells of interest in a complex heterogeneous tissue section, under direct microscopic visualization. Recently, there has been a surge of publications using LCM for tissue spatial molecular profiling relevant to a wide range of research topics. AREAS COVERED: We summarize the many advances in tissue Laser Capture Proteomics (LCP) using mass spectrometry for discovery, and protein arrays for signal pathway network mapping. This review emphasizes: a) transition of LCM phosphoproteomics from the lab to the clinic for individualized cancer therapy, and b) the emerging frontier of LCM single cell molecular analysis combining proteomics with genomic, and transcriptomic analysis. The search strategy was based on the combination of MeSH terms with expert refinement. EXPERT OPINION: LCM is complemented by a rich set of instruments, methodology protocols, and analytical A.I. (artificial intelligence) software for basic and translational research. Resolution is advancing to the tissue single cell level. A vision for the future evolution of LCM is presented. Emerging LCM technology is combining digital and AI guided remote imaging with automation, and telepathology, to a achieve multi-omic profiling that was not previously possible.


Asunto(s)
Medicina de Precisión , Proteómica , Inteligencia Artificial , Captura por Microdisección con Láser , Rayos Láser
9.
J Biol Chem ; 294(29): 11180-11198, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31167787

RESUMEN

Protein-protein interactions lie at the heart of many biological processes and therefore represent promising drug targets. Despite this opportunity, identification of protein-protein interfaces remains challenging. We have previously developed a method that relies on coating protein surfaces with small-molecule dyes to discriminate between solvent-accessible protein surfaces and hidden interface regions. Dye-bound, solvent-accessible protein regions resist trypsin digestion, whereas hidden interface regions are revealed by denaturation and sequenced by MS. The small-molecule dyes bind promiscuously and with high affinity, but their binding mechanism is unknown. Here, we report on the optimization of a novel dye probe used in protein painting, Fast Blue B + naphthionic acid, and show that its affinity for proteins strongly depends on hydrophobic moieties that we call here "hydrophobic clamps." We demonstrate the utility of this probe by sequencing the protein-protein interaction regions between the Hippo pathway protein Yes-associated protein 2 (YAP2) and tight junction protein 1 (TJP1 or ZO-1), uncovering interactions via the known binding domain as well as ZO-1's MAGUK domain and YAP's N-terminal proline-rich domain. Additionally, we demonstrate how residues predicted by protein painting are present exclusively in the complex interface and how these residues may guide the development of peptide inhibitors using a case study of programmed cell death protein 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1). Inhibitors designed around the PD-1/PD-L1 interface regions identified via protein painting effectively disrupted complex formation, with the most potent inhibitor having an IC50 of 5 µm.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígeno B7-H1/metabolismo , Espectrometría de Masas/métodos , Receptor de Muerte Celular Programada 1/metabolismo , Factores de Transcripción/metabolismo , Proteína de la Zonula Occludens-1/metabolismo , Aminoácidos/metabolismo , Compuestos Azo/metabolismo , Sitios de Unión , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Unión Proteica , Proteínas Señalizadoras YAP
10.
Breast Cancer Res ; 22(1): 135, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33267867

RESUMEN

BACKGROUND: The lack of specificity and high degree of false positive and false negative rates when using mammographic screening for detecting early-stage breast cancer is a critical issue. Blood-based molecular assays that could be used in adjunct with mammography for increased specificity and sensitivity could have profound clinical impact. Our objective was to discover and independently verify a panel of candidate blood-based biomarkers that could identify the earliest stages of breast cancer and complement current mammographic screening approaches. METHODS: We used affinity hydrogel nanoparticles coupled with LC-MS/MS analysis to enrich and analyze low-abundance proteins in serum samples from 20 patients with invasive ductal carcinoma (IDC) breast cancer and 20 female control individuals with positive mammograms and benign pathology at biopsy. We compared these results to those obtained from five cohorts of individuals diagnosed with cancer in organs other than breast (ovarian, lung, prostate, and colon cancer, as well as melanoma) to establish IDC-specific protein signatures. Twenty-four IDC candidate biomarkers were then verified by multiple reaction monitoring (LC-MRM) in an independent validation cohort of 60 serum samples specifically including earliest-stage breast cancer and benign controls (19 early-stage (T1a) IDC and 41 controls). RESULTS: In our discovery set, 56 proteins were increased in the serum samples from IDC patients, and 32 of these proteins were specific to IDC. Verification of a subset of these proteins in an independent cohort of early-stage T1a breast cancer yielded a panel of 4 proteins, ITGA2B (integrin subunit alpha IIb), FLNA (Filamin A), RAP1A (Ras-associated protein-1A), and TLN-1 (Talin-1), which classified breast cancer patients with 100% sensitivity and 85% specificity (AUC of 0.93). CONCLUSIONS: Using a nanoparticle-based protein enrichment technology, we identified and verified a highly specific and sensitive protein signature indicative of early-stage breast cancer with no false positives when assessing benign and inflammatory controls. These markers have been previously reported in cell-ECM interaction and tumor microenvironment biology. Further studies with larger cohorts are needed to evaluate whether this biomarker panel improves the positive predictive value of mammography for breast cancer detection.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias de la Mama/diagnóstico , Carcinoma Ductal de Mama/diagnóstico , Detección Precoz del Cáncer/métodos , Proteínas de la Matriz Extracelular/sangre , Adulto , Anciano , Biopsia , Mama/diagnóstico por imagen , Mama/patología , Neoplasias de la Mama/sangre , Carcinoma Ductal de Mama/sangre , Carcinoma Ductal de Mama/patología , Estudios de Casos y Controles , Estudios de Cohortes , Proteínas de la Matriz Extracelular/química , Femenino , Humanos , Masculino , Mamografía , Persona de Mediana Edad , Nanopartículas/química , Proteómica/métodos
11.
Parasitol Res ; 119(7): 2227-2235, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32435898

RESUMEN

Babesiosis among humans is on the rise in North America. Current diagnostic assays for the screening of babesiosis require blood collection by venipuncture, which is an invasive method. Urine on the other hand is a desirable biospecimen for biomarker analysis of Babesia microti infections because it can be collected periodically and non-invasively. Our group uses a new class of biomarker harvesting nanocage technology, which, when combined with mass spectrometry (MS), can determine the presence of parasite proteins shed in different bodily fluids of mammalian hosts, including urine. Using the hamster model of babesiosis, our nanoparticle-MS approach identified several B. microti proteins in erythrocytes, plasma, and urine samples. Surface and secreted antigens previously shown to elicit host immune responses against the parasite were particularly abundant in erythrocytes and plasma compared to other proteins. Two of these antigens, BmSA1 and BMR1_03g00947, showed different localization patterns by immunofluorescence of infected erythrocytes. Hamster urine samples from parasitemic animals harbored lower numbers of B. microti proteins compared to erythrocytes and plasma, with glycolytic enzymes, cytoskeletal components, and chaperones being the most frequently detected proteins. By applying novel nanoparticle-MS methods, a high level of analytical sensitivity can be achieved to detect multiple B. microti proteins in blood and urine. This is generally difficult to obtain with other techniques due to the masking of parasite biomarkers by the complex biomolecular matrix of bodily fluids from the host.


Asunto(s)
Babesia microti/aislamiento & purificación , Babesiosis/diagnóstico , Eritrocitos/parasitología , Proteínas Protozoarias/metabolismo , Animales , Babesia microti/metabolismo , Babesiosis/sangre , Babesiosis/orina , Biomarcadores/sangre , Biomarcadores/metabolismo , Biomarcadores/orina , Cricetinae , Espectrometría de Masas , Proteómica , Proteínas Protozoarias/sangre , Proteínas Protozoarias/orina , Sensibilidad y Especificidad
12.
Clin Infect Dis ; 68(6): 1052-1057, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30307486

RESUMEN

Borrelia burgdorferi was discovered to be the cause of Lyme disease in 1983, leading to seroassays. The 1994 serodiagnostic testing guidelines predated a full understanding of key B. burgdorferi antigens and have a number of shortcomings. These serologic tests cannot distinguish active infection, past infection, or reinfection. Reliable direct-detection methods for active B. burgdorferi infection have been lacking in the past but are needed and appear achievable. New approaches have effectively been applied to other emerging infections and show promise in direct detection of B. burgdorferi infections.


Asunto(s)
Borrelia burgdorferi , Enfermedad de Lyme/diagnóstico , Enfermedad de Lyme/microbiología , Borrelia burgdorferi/genética , Pruebas Diagnósticas de Rutina , Genómica/métodos , Ensayos Analíticos de Alto Rendimiento , Humanos , Reacción en Cadena de la Polimerasa , Pruebas Serológicas
13.
Lab Invest ; 99(5): 708-721, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30659273

RESUMEN

Bone tissue is critically lagging behind soft tissues and biofluids in our effort to advance precision medicine. The main challenges have been accessibility and the requirement for deleterious decalcification processes that impact the fidelity of diagnostic histomorphology and hinder downstream analyses such as fluorescence in-situ hybridization (FISH). We have developed an alternative fixation chemistry that simultaneously fixes and decalcifies bone tissue. We compared tissue morphology, immunohistochemistry (IHC), cell signal phosphoprotein analysis, and FISH in 50 patient matched primary bone cancer cases that were either formalin fixed and decalcified, or theralin fixed with and without decalcification. Use of theralin improved tissue histomorphology, whereas overall IHC was comparable to formalin fixed, decalcified samples. Theralin-fixed samples showed a significant increase in protein and DNA extractability, supporting technologies such as laser-capture microdissection and reverse phase protein microarrays. Formalin-fixed bone samples suffered from a fixation artifact where protein quantification of ß-actin directly correlated with fixation time. Theralin-fixed samples were not affected by this artifact. Moreover, theralin fixation enabled standard FISH staining in bone cancer samples, whereas no FISH staining was observed in formalin-fixed samples. We conclude that the use of theralin fixation unlocks the molecular archive within bone tissue allowing bone to enter the standard tissue analysis pipeline. This will have significant implications for bone cancer patients, in whom personalized medicine has yet to be implemented.


Asunto(s)
Huesos/metabolismo , Expresión Génica , Hibridación Fluorescente in Situ/métodos , Proteoma/metabolismo , Proteómica/métodos , Animales , Neoplasias Óseas/diagnóstico , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Huesos/patología , Fijadores/química , Formaldehído/química , Humanos , Inmunohistoquímica , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Reproducibilidad de los Resultados , Fijación del Tejido/métodos
14.
J Virol ; 92(15)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29769351

RESUMEN

Protein phosphatase 1 (PP1) is a serine/threonine phosphatase which has been implicated in the regulation of a number of viruses, including HIV-1, Ebolavirus, and Rift Valley fever virus. Catalytic subunits of PP1 (PP1α, PP1ß, and PP1γ) interact with a host of regulatory subunits and target a wide variety of cellular substrates through a combination of short binding motifs, including an RVxF motif present in the majority of PP1 regulatory subunits. Targeting the RVxF-interacting site on PP1 with the small molecule 1E7-03 inhibits HIV-1, Ebolavirus, and Rift Valley fever virus replication. In this study, we determined the effect of PP1 on Venezuelan equine encephalitis virus (VEEV) replication. Treatment of VEEV-infected cells with 1E7-03 decreased viral replication by more than 2 logs (50% effective concentration [EC50] = 0.6 µM). 1E7-03 treatment reduced viral titers starting at 8 h postinfection. Viral replication was also decreased after treatment with PP1α-targeting small interfering RNA (siRNA). Confocal microscopy demonstrated that PP1α shuttles toward the cytosol during infection with VEEV and that PP1α colocalizes with VEEV capsid. Coimmunoprecipitation experiments confirmed VEEV capsid interaction with PP1α. Furthermore, immunoprecipitation and mass spectrometry data showed that VEEV capsid is phosphorylated and that phosphorylation is moderated by PP1α. Finally, less viral RNA is associated with capsid after treatment with 1E7-03. Coupled with data showing that 1E7-03 inhibits several alphaviruses, this study indicates that inhibition of the PP1α RVxF binding pocket is a promising therapeutic target and provides novel evidence that PP1α modulation of VEEV capsid phosphorylation influences viral replication.IMPORTANCE Venezuelan equine encephalitis virus (VEEV) causes moderate flu-like symptoms and can lead to severe encephalitic disease and potentially death. There are currently no FDA-approved therapeutics or vaccines for human use, and understanding the molecular underpinning of host-virus interactions can aid in the rational design of intervention strategies. The significance of our research is in identifying the interaction between protein phosphatase 1 (PP1) and the viral capsid protein. This interaction is important for viral replication, as inhibition of PP1 results in decrease viral replication. Inhibition of PP1 also inhibited multiple biomedically important alphaviruses, indicating that PP1 may be a potential therapeutic target for alphavirus-induced disease.


Asunto(s)
Proteínas de la Cápside/metabolismo , Cápside/metabolismo , Virus de la Encefalitis Equina Venezolana/fisiología , Proteína Fosfatasa 1/metabolismo , Replicación Viral/fisiología , Animales , Proteínas de la Cápside/genética , Chlorocebus aethiops , Fosforilación/genética , Proteína Fosfatasa 1/genética , Células Vero
16.
Cancer Treat Res ; 178: 171-187, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31209845

RESUMEN

Genomic analysis of tumor specimens has revealed that cancer is fundamentally a proteomic disease at the functional level: driven by genomically defined derangements, but selected for in the proteins that are encoded and the aberrant activation of signaling and biochemical networks. This activation is measured by posttranslational modifications such as phosphorylation and other modifications that modulate cellular signaling, and these events cannot be effectively measured by genomic analysis alone. Moreover, these signaling networks by and large represent the targets for many FDA-approved and experimental molecularly targeted therapeutics. Consequently, it is important that we consider new classification schemas for oncology based not on tumor site of origin or histology under the microscope but on the functional protein signaling architecture. There are numerous proteomic technologies that could be discussed from a purely technological standpoint, but this chapter will concentrate on an overview of the main proteomic technologies available for conducting protein pathway activation analysis of clinical specimens such as multiplex immunoassays, phospho-specific flow cytometry, reverse phase protein microarrays, quantitative immunohistochemistry, and mass spectrometry. This chapter will focus on the application of these technologies to cancer-based clinical studies evaluating prognostic/predictive markers or for stratifying patients to personalized treatments.


Asunto(s)
Neoplasias , Medicina de Precisión , Proteómica , Humanos , Neoplasias/genética , Neoplasias/terapia , Análisis por Matrices de Proteínas , Transducción de Señal
17.
Adv Exp Med Biol ; 1188: 95-111, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31820385

RESUMEN

Reverse phase protein microarrays (RPPA) and laser capture microdissection (LCM) are "sibling" technologies that originated from the same laboratory to overcome the challenge of quantifying low-abundance proteins in heterogeneous tissues. Combining both technologies provides both unique opportunities and unique challenges. Enabling the unprecedented resolution of the activation state of labile biomarkers, such as phosphorylated cell signaling proteins, has had a substantial impact on our understanding of diseases and is playing a significant role in clinical trials. At the same time, quantifying proteins at this sensitivity in very small amounts of material requires cognizance of pre-analytical variability and the limits of downstream detection technologies. Here, we discuss both the potential that the combination of both technologies presents and the potential pitfalls that must be navigated.


Asunto(s)
Captura por Microdisección con Láser , Análisis por Matrices de Proteínas , Proteínas , Análisis por Matrices de Proteínas/métodos , Análisis por Matrices de Proteínas/normas , Análisis por Matrices de Proteínas/tendencias , Proteínas/química , Tecnología/tendencias
18.
Adv Exp Med Biol ; 1188: 1-19, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31820380

RESUMEN

RPPA technology has graduated from a research tool to an essential component of clinical drug discovery research and personalized medicine. Next generations of RPPA technology will be a single clinical instrument that integrates all the steps of the workflow.


Asunto(s)
Medicina de Precisión , Análisis por Matrices de Proteínas , Proteómica , Medicina de Precisión/instrumentación , Medicina de Precisión/tendencias , Análisis por Matrices de Proteínas/normas , Análisis por Matrices de Proteínas/tendencias , Investigación/instrumentación , Investigación/tendencias
19.
Proc Natl Acad Sci U S A ; 113(3): E282-90, 2016 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-26729871

RESUMEN

In the lactating mammary gland, the plasma membrane calcium ATPase2 (PMCA2) transports milk calcium. Its expression is activated in breast cancers, where high tumor levels predict increased mortality. We find that PMCA2 expression correlates with HER2 levels in breast cancers and that PMCA2 interacts with HER2 in specific actin-rich membrane domains. Knocking down PMCA2 increases intracellular calcium, disrupts interactions between HER2 and HSP-90, inhibits HER2 signaling, and results in internalization and degradation of HER2. Manipulating PMCA2 levels regulates the growth of breast cancer cells, and knocking out PMCA2 inhibits the formation of tumors in mouse mammary tumor virus (MMTV)-Neu mice. These data reveal previously unappreciated molecular interactions regulating HER2 localization, membrane retention, and signaling, as well as the ability of HER2 to generate breast tumors, suggesting that interactions between PMCA2 and HER2 may represent therapeutic targets for breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Receptor ErbB-2/metabolismo , Transducción de Señal , Animales , Neoplasias de la Mama/patología , Calcio/farmacología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Endocitosis/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Immunoblotting , Espacio Intracelular/metabolismo , Neoplasias Mamarias Animales , Ratones , Unión Proteica , Transporte de Proteínas , Análisis de Supervivencia
20.
J Infect Dis ; 218(suppl_5): S365-S387, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30169850

RESUMEN

Background: Ebola virus (EBOV) mainly targets myeloid cells; however, extensive death of T cells is often observed in lethal infections. We have previously shown that EBOV VP40 in exosomes causes recipient immune cell death. Methods: Using VP40-producing clones, we analyzed donor cell cycle, extracellular vesicle (EV) biogenesis, and recipient immune cell death. Transcription of cyclin D1 and nuclear localization of VP40 were examined via kinase and chromatin immunoprecipitation assays. Extracellular vesicle contents were characterized by mass spectrometry, cytokine array, and western blot. Biosafety level-4 facilities were used for wild-type Ebola virus infection studies. Results: VP40 EVs induced apoptosis in recipient T cells and monocytes. VP40 clones were accelerated in growth due to cyclin D1 upregulation, and nuclear VP40 was found bound to the cyclin D1 promoter. Accelerated cell cycling was related to EV biogenesis, resulting in fewer but larger EVs. VP40 EV contents were enriched in ribonucleic acid-binding proteins and cytokines (interleukin-15, transforming growth factor-ß1, and interferon-γ). Finally, EBOV-infected cell and animal EVs contained VP40, nucleoprotein, and glycoprotein. Conclusions: Nuclear VP40 upregulates cyclin D1 levels, resulting in dysregulated cell cycle and EV biogenesis. Packaging of cytokines and EBOV proteins into EVs from infected cells may be responsible for the decimation of immune cells during EBOV pathogenesis.


Asunto(s)
Ciclo Celular/fisiología , Ebolavirus/metabolismo , Vesículas Extracelulares/metabolismo , Fiebre Hemorrágica Ebola/metabolismo , Fiebre Hemorrágica Ebola/virología , Nucleoproteínas/metabolismo , Proteínas del Núcleo Viral/metabolismo , Apoptosis/fisiología , Línea Celular , Línea Celular Tumoral , Ciclina D1/metabolismo , Exosomas/metabolismo , Vesículas Extracelulares/virología , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Regiones Promotoras Genéticas/fisiología , Unión Proteica/fisiología , Células U937 , Regulación hacia Arriba/fisiología , Proteínas de la Matriz Viral/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA