Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Biomark Res ; 11(1): 73, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37491309

ABSTRACT

BACKGROUND: A subset of triple-negative breast cancers (TNBCs) have homologous recombination deficiency with upregulation of compensatory DNA repair pathways. PIKTOR, a combination of TAK-228 (TORC1/2 inhibitor) and TAK-117 (PI3Kα inhibitor), is hypothesized to increase genomic instability and increase DNA damage repair (DDR) deficiency, leading to increased sensitivity to DNA-damaging chemotherapy and to immune checkpoint blockade inhibitors. METHODS: 10 metastatic TNBC patients received 4 mg TAK-228 and 200 mg TAK-117 (PIKTOR) orally each day for 3 days followed by 4 days off, weekly, until disease progression (PD), followed by intravenous cisplatin 75 mg/m2 plus nab paclitaxel 220 mg/m2 every 3 weeks for up to 6 cycles. Patients received subsequent treatment with pembrolizumab and/or chemotherapy. Primary endpoints were objective response rate with cisplatin/nab paclitaxel and safety. Biopsies of a metastatic lesion were collected prior to and at PD on PIKTOR. Whole exome and RNA-sequencing and reverse phase protein arrays (RPPA) were used to phenotype tumors pre- and post-PIKTOR for alterations in DDR, proliferation, and immune response. RESULTS: With cisplatin/nab paclitaxel (cis/nab pac) therapy post PIKTOR, 3 patients had clinical benefit (1 partial response (PR) and 2 stable disease (SD) ≥ 6 months) and continued to have durable benefit in progression-free survival with pembrolizumab post-cis/nab pac for 1.2, 2, and 3.6 years. Their post-PIKTOR metastatic tissue displayed decreased mismatch repair (MMR), increased tumor mutation burden, and significantly lower levels of 53BP1, DAG Lipase ß, GCN2, AKT Ser473, and PKCzeta Thr410/403 compared to pre-PIKTOR tumor tissue. CONCLUSIONS: Priming patients' chemotherapy-pretreated metastatic TNBC with PIKTOR led to very prolonged response/disease control with subsequent cis/nab pac, followed by pembrolizumab, in 3 of 10 treated patients. Our multi-omics approach revealed a higher number of genomic alterations, reductions in MMR, and alterations in immune and stress response pathways post-PIKTOR in patients who had durable responses. TRIAL REGISTRATION: This clinical trial was registered on June 21, 2017, at ClinicalTrials.gov using identifier NCT03193853.

2.
J Appl Stat ; 49(16): 4278-4293, 2022.
Article in English | MEDLINE | ID: mdl-36353301

ABSTRACT

In disease screening, a biomarker combination developed by combining multiple markers tends to have a higher sensitivity than an individual marker. Parametric methods for marker combination rely on the inverse of covariance matrices, which is often a non-trivial problem for high-dimensional data generated by modern high-throughput technologies. Additionally, another common problem in disease diagnosis is the existence of limit of detection (LOD) for an instrument - that is, when a biomarker's value falls below the limit, it cannot be observed and is assigned an NA value. To handle these two challenges in combining high-dimensional biomarkers with the presence of LOD, we propose a resample-replace lasso procedure. We first impute the values below LOD and then use the graphical lasso method to estimate the means and precision matrices for the high-dimensional biomarkers. The simulation results show that our method outperforms alternative methods such as either substitute NA values with LOD values or remove observations that have NA values. A real case analysis on a protein profiling study of glioblastoma patients on their survival status indicates that the biomarker combination obtained through the proposed method is more accurate in distinguishing between two groups.

3.
Med ; 2(6): 736-754, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34223403

ABSTRACT

BACKGROUND: Upregulated glucose metabolism is a common feature of tumors. Glucose can be broken down by either glycolysis or the oxidative pentose phosphate pathway (oxPPP). The relative usage within tumors of these catabolic pathways remains unclear. Similarly, the extent to which tumors make biomass precursors from glucose, versus take them up from the circulation, is incompletely defined. METHODS: We explore human triple negative breast cancer (TNBC) metabolism by isotope tracing with [1,2-13C]glucose, a tracer that differentiates glycolytic versus oxPPP catabolism and reveals glucose-driven anabolism. Patients enrolled in clinical trial NCT03457779 and received IV infusion of [1,2-13C]glucose during core biopsy of their primary TNBC. Tumor samples were analyzed for metabolite labeling by liquid chromatography-mass spectrometry (LC-MS). Genomic and proteomic analyses were performed and related to observed metabolic fluxes. FINDINGS: TNBC ferments glucose to lactate, with glycolysis dominant over the oxPPP. Most ribose phosphate is nevertheless produced by oxPPP. Glucose also feeds amino acid synthesis, including of serine, glycine, aspartate, glutamate, proline and glutamine (but not asparagine). Downstream in glycolysis, tumor pyruvate and lactate labeling exceeds that found in serum, indicating that lactate exchange via monocarboxylic transporters is less prevalent in human TNBC compared with most normal tissues or non-small cell lung cancer. CONCLUSIONS: Glucose directly feeds ribose phosphate, amino acid synthesis, lactate, and the TCA cycle locally within human breast tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Triple Negative Breast Neoplasms , Amino Acids , Glucose/metabolism , Humans , Lactic Acid/metabolism , Proteomics , Ribosemonophosphates
4.
Cureus ; 13(2): e13499, 2021 Feb 22.
Article in English | MEDLINE | ID: mdl-33777584

ABSTRACT

Thrombus characteristics are dependent on clot composition, but identification of the etiology based on histological analysis has proved inconclusive. Identification of proteomic signatures may help to differentiate between clots of different etiologies such as cardioembolic, large artery atherosclerotic, and other known etiologies, information that could enhance an individualized medicine approach to secondary stroke prevention. In this study, total protein extracts from cardioembolic (n=25) and large artery atherosclerotic (n=23) thrombus specimens were arrayed in quadruplicate on nitrocellulose slides and immunostained for 31 proteins using a Dako Autostainer (Agilent Technologies, Inc., Santa Clara, USA). We quantified 31 proteins involved in platelet and/or endothelial function, inflammation, oxidative stress, and metabolism. Pathway analysis showed more heterogeneity and protein network interactions in the cardioembolic clots but no specific correlations with clot etiology. Reverse-phase protein arrays are a powerful tool for assessing cellular interactions within the clot microenvironment and may enhance understanding of clot formation and origination. This tool could be further explored to help in identifying stroke etiology in large vessel occlusion patients with embolic stroke of an undetermined source.

5.
J Infect Dis ; 223(10): 1677-1680, 2021 05 28.
Article in English | MEDLINE | ID: mdl-33718952

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19/immunology , SARS-CoV-2/immunology , Asymptomatic Diseases , COVID-19/epidemiology , COVID-19 Nucleic Acid Testing , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Health Personnel , Humans , Male , Neutralization Tests , Surveys and Questionnaires , Time Factors , Virginia/epidemiology
6.
Sci Rep ; 10(1): 19340, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33168903

ABSTRACT

Mass spectrometry enhanced by nanotechnology can achieve previously unattainable sensitivity for characterizing urinary pathogen-derived peptides. We utilized mass spectrometry enhanced by affinity hydrogel particles (analytical sensitivity = 2.5 pg/mL) to study tick pathogen-specific proteins shed in the urine of patients with (1) erythema migrans rash and acute symptoms, (2) post treatment Lyme disease syndrome (PTLDS), and (3) clinical suspicion of tick-borne illnesses (TBI). Targeted pathogens were Borrelia, Babesia, Anaplasma, Rickettsia, Ehrlichia, Bartonella, Francisella, Powassan virus, tick-borne encephalitis virus, and Colorado tick fever virus. Specificity was defined by 100% amino acid sequence identity with tick-borne pathogen proteins, evolutionary taxonomic verification for related pathogens, and no identity with human or other organisms. Using a cut off of two pathogen peptides, 9/10 acute Lyme Borreliosis patients resulted positive, while we identified zero false positive in 250 controls. Two or more pathogen peptides were identified in 40% of samples from PTLDS and TBI patients (categories 2 and 3 above, n = 59/148). Collectively, 279 distinct unique tick-borne pathogen derived peptides were identified. The number of pathogen specific peptides was directly correlated with presence or absence of symptoms reported by patients (ordinal regression pseudo-R2 = 0.392, p = 0.010). Enhanced mass spectrometry is a new tool for studying tick-borne pathogen infections.


Subject(s)
Lyme Disease/microbiology , Lyme Disease/urine , Peptides/urine , Ticks , Adult , Aged , Algorithms , Animals , Babesia microti/metabolism , Biomarkers/metabolism , Borrelia , Erythema Chronicum Migrans/microbiology , Erythema Chronicum Migrans/urine , Exanthema , Female , Humans , Hydrogels/chemistry , Infectious Disease Medicine , Male , Mass Spectrometry , Mesocricetus , Middle Aged , Peptides/chemistry , Regression Analysis , Urinalysis
7.
medRxiv ; 2020 Aug 02.
Article in English | MEDLINE | ID: mdl-32766594

ABSTRACT

Coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), became a pandemic in early 2020. Lateral flow immunoassays for antibody testing have been viewed as a cheap and rapidly deployable method for determining previous infection with SARS-CoV-2; however, these assays have shown unacceptably low sensitivity. We report on nine lateral flow immunoassays currently available and compare their titer sensitivity in serum to a best-practice enzyme-linked immunosorbent assay (ELISA) and viral neutralization assay. For a small group of PCR-positive, we found two lateral flow immunoassay devices with titer sensitivity roughly equal to the ELISA; these devices were positive for all PCR-positive patients harboring SARS-CoV-2 neutralizing antibodies. One of these devices was deployed in Northern Italy to test its sensitivity and specificity in a real-world clinical setting. Using the device with fingerstick blood on a cohort of 27 hospitalized PCR-positive patients and seven hospitalized controls, ROC curve analysis gave AUC values of 0.7646 for IgG. For comparison, this assay was also tested with saliva from the same patient population and showed reduced discrimination between cases and controls with AUC values of 0.6841 for IgG. Furthermore, during viral neutralization testing, one patient was discovered to harbor autoantibodies to ACE2, with implications for how immune responses are profiled. We show here through a proof-of-concept study that these lateral flow devices can be as analytically sensitive as ELISAs and adopted into hospital protocols; however, additional improvements to these devices remain necessary before their clinical deployment.

8.
Adv Exp Med Biol ; 1188: 61-75, 2019.
Article in English | MEDLINE | ID: mdl-31820383

ABSTRACT

Reverse phase protein arrays (RPPA) are miniature dot blots constructed using robotic arrayers to deposit protein containing samples onto nitrocellulose-coated glass slides. Reverse phase protein arrays address the challenge of quantifying low-abundance proteins and posttranslationally modified proteins in cellular lysates and body fluids. RPPA technology is ideally suited to biomarker discovery, signal pathway profiling, functional phenotype analysis, and mechanism of action studies for drug discovery. Each array is fabricated with specimens, controls, and calibrators, thus providing a complete assay on each slide. Constructing a reverse phase protein array initially consists of selecting an arrayer, pin type, print head configuration, and nitrocellulose slide that is optimized for the particular specimen type and protein detection method. Herein we present the nuances of RPPA fabrication and study design using a solid pin arrayer and nitrocellulose-coated slides.


Subject(s)
Protein Array Analysis , Proteins , Collodion , Drug Discovery/instrumentation , Drug Discovery/trends , Printing, Three-Dimensional , Protein Array Analysis/instrumentation , Proteins/chemistry
9.
Adv Exp Med Biol ; 1188: 95-111, 2019.
Article in English | MEDLINE | ID: mdl-31820385

ABSTRACT

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.


Subject(s)
Laser Capture Microdissection , Protein Array Analysis , Proteins , Protein Array Analysis/methods , Protein Array Analysis/standards , Protein Array Analysis/trends , Proteins/chemistry , Technology/trends
10.
Sci Rep ; 9(1): 17380, 2019 11 22.
Article in English | MEDLINE | ID: mdl-31758030

ABSTRACT

Tumor clonal heterogeneity drives treatment resistance. But robust models are lacking that permit eavesdropping on the basic interaction network of tumor clones. We developed an in vitro, functional model of clonal cooperation using U87MG glioblastoma cells, which isolates fundamental clonal interactions. In this model pre-labeled clones are co-cultured to track changes in their individual motility, growth, and drug resistance behavior while mixed. This highly reproducible system allowed us to address a new class of fundamental questions about clonal interactions. We demonstrate that (i) a single clone can switch off the motility of the entire multiclonal U87MG cell line in 3D culture, (ii) maintenance of clonal heterogeneity is an intrinsic and influential cancer cell property, where clones coordinate growth rates to protect slow growing clones, and (iii) two drug sensitive clones can develop resistance de novo when cooperating. Furthermore, clonal communication for these specific types of interaction did not require diffusible factors, but appears to depend on cell-cell contact. This model constitutes a straightforward but highly reliable tool for isolating the complex clonal interactions that make up the fundamental "hive mind" of the tumor. It uniquely exposes clonal interactions for future pharmacological and biochemical studies.


Subject(s)
Brain Neoplasms/pathology , Cell Movement , Cell Proliferation , Clone Cells/pathology , Drug Resistance, Neoplasm , Glioblastoma/pathology , Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Communication/drug effects , Cell Communication/physiology , Cell Line, Tumor , Clonal Evolution/physiology , Clone Cells/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Genetic Heterogeneity , Genotype , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Models, Biological , Signal Transduction/genetics
11.
Lab Invest ; 99(5): 708-721, 2019 05.
Article in English | MEDLINE | ID: mdl-30659273

ABSTRACT

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.


Subject(s)
Bone and Bones/metabolism , Gene Expression , In Situ Hybridization, Fluorescence/methods , Proteome/metabolism , Proteomics/methods , Animals , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone and Bones/pathology , Fixatives/chemistry , Formaldehyde/chemistry , Humans , Immunohistochemistry , Mice, Inbred BALB C , Mice, Inbred ICR , Reproducibility of Results , Tissue Fixation/methods
12.
Nat Genet ; 50(5): 708-717, 2018 05.
Article in English | MEDLINE | ID: mdl-29686388

ABSTRACT

To understand how genomic heterogeneity of glioblastoma (GBM) contributes to poor therapy response, we performed DNA and RNA sequencing on GBM samples and the neurospheres and orthotopic xenograft models derived from them. We used the resulting dataset to show that somatic driver alterations including single-nucleotide variants, focal DNA alterations and oncogene amplification on extrachromosomal DNA (ecDNA) elements were in majority propagated from tumor to model systems. In several instances, ecDNAs and chromosomal alterations demonstrated divergent inheritance patterns and clonal selection dynamics during cell culture and xenografting. We infer that ecDNA was unevenly inherited by offspring cells, a characteristic that affects the oncogenic potential of cells with more or fewer ecDNAs. Longitudinal patient tumor profiling found that oncogenic ecDNAs are frequently retained throughout the course of disease. Our analysis shows that extrachromosomal elements allow rapid increase of genomic heterogeneity during GBM evolution, independently of chromosomal DNA alterations.


Subject(s)
Brain Neoplasms/genetics , DNA, Neoplasm/genetics , Glioblastoma/genetics , Animals , Cell Line, Tumor , Chromosomes , Female , Genomics/methods , Heredity , Humans , Mice , Mice, Nude , Oncogenes , Polymorphism, Single Nucleotide
13.
Expert Rev Proteomics ; 15(2): 131-152, 2018 02.
Article in English | MEDLINE | ID: mdl-29271260

ABSTRACT

INTRODUCTION: Breast cancer subtypes are currently defined by a combination of morphologic, genomic, and proteomic characteristics. These subtypes provide a molecular portrait of the tumor that aids diagnosis, prognosis, and treatment escalation/de-escalation options. Gene expression signatures describing intrinsic breast cancer subtypes for predicting risk of recurrence have been rapidly adopted in the clinic. Despite the use of subtype classifications, many patients develop drug resistance, breast cancer recurrence, or therapy failure. Areas covered: This review provides a summary of immunohistochemistry, reverse phase protein array, mass spectrometry, and integrative studies that are revealing differences in biological functions within and between breast cancer subtypes. We conclude with a discussion of rigor and reproducibility for proteomic-based biomarker discovery. Expert commentary: Innovations in proteomics, including implementation of assay guidelines and standards, are facilitating refinement of breast cancer subtypes. Proteomic and phosphoproteomic information distinguish biologically functional subtypes, are predictive of recurrence, and indicate likelihood of drug resistance. Actionable, activated signal transduction pathways can now be quantified and characterized. Proteomic biomarker validation in large, well-designed studies should become a public health priority to capitalize on the wealth of information gleaned from the proteome.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Proteomics/methods , Breast Neoplasms/classification , Breast Neoplasms/diagnosis , Female , Humans , Mass Spectrometry/methods
14.
Methods Mol Biol ; 1606: 85-102, 2017.
Article in English | MEDLINE | ID: mdl-28501995

ABSTRACT

Bone metastasis from primary cancer sites creates diagnostic and therapeutic challenges. Calcified bone is difficult to biopsy due to tissue hardness and patient discomfort, thus limiting the frequency and availability of bone/bone marrow biopsy material for molecular profiling. In addition, bony tissue must be demineralized (decalcified) prior to histomorphologic analysis. Decalcification processes rely on three main principles: (a) solubility of calcium salts in an acid, such as formic or nitric acid; (b) calcium chelation with ethylenediaminetetraacetic acid (EDTA); or (c) ion-exchange resins in a weak acid. A major roadblock in molecular profiling of bony tissue has been the lack of a suitable demineralization process that preserves histomorphology of calcified and soft tissue elements while also preserving phosphoproteins and nucleic acids. In this chapter, we describe general issues relevant to specimen collection and preservation of osseous tissue for molecular profiling. We provide two protocols: (a) one-step preservation of tissue histomorphology and proteins and posttranslational modifications, with simultaneous decalcification of bony tissue, and (b) ethanol-based tissue processing for TheraLin-fixed bony tissue.


Subject(s)
Bone and Bones/anatomy & histology , Decalcification Technique/methods , Preservation, Biological/methods , Animals , Bone Neoplasms/diagnosis , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone and Bones/metabolism , Bone and Bones/pathology , Humans
15.
Methods Mol Biol ; 1606: 115-132, 2017.
Article in English | MEDLINE | ID: mdl-28501997

ABSTRACT

Laser capture microdissection (LCM) is a technique that allows procurement of an enriched cell population from a heterogeneous tissue sample under direct microscopic visualization. Fundamentally, laser capture microdissection consists of three main steps: (1) visualizing the desired cell population by microscopy, (2) melting a thermolabile polymer onto the desired cell populations using infrared laser energy to form a polymer-cell composite (capture method) or photovolatizing a region of tissue using ultraviolet laser energy (cutting method), and (3) removing the desired cell population from the heterogeneous tissue. In this chapter, we discuss the infrared capture method only. LCM technology is compatible with a wide range of downstream applications such as mass spectrometry, DNA genotyping and RNA transcript profiling, cDNA library generation, proteomics discovery, and signal pathway mapping. This chapter profiles the ArcturusXT™ laser capture microdissection instrument, using isolation of specific cortical lamina from nonhuman primate brain regions, and sample preparation methods for downstream proteomic applications.


Subject(s)
Brain/anatomy & histology , Laser Capture Microdissection/methods , Primates/anatomy & histology , Proteomics/methods , Specimen Handling/methods , Animals , Brain/metabolism , Laser Capture Microdissection/instrumentation , Primates/metabolism
16.
Antiviral Res ; 143: 162-175, 2017 07.
Article in English | MEDLINE | ID: mdl-28442428

ABSTRACT

Despite over 60 years of research on antiviral drugs, very few are FDA approved to treat acute viral infections. Rift Valley fever virus (RVFV), an arthropod borne virus that causes hemorrhagic fever in severe cases, currently lacks effective treatments. Existing as obligate intracellular parasites, viruses have evolved to manipulate host cell signaling pathways to meet their replication needs. Specifically, translation modulation is often necessary for viruses to establish infection in their host. Here we demonstrated phosphorylation of p70 S6 kinase, S6 ribosomal protein, and eIF4G following RVFV infection in vitro through western blot analysis and in a mouse model of infection through reverse phase protein microarrays (RPPA). Inhibition of p70 S6 kinase through rapamycin treatment reduced viral titers in vitro and increased survival and mitigated clinical disease in RVFV challenged mice. Additionally, the phosphorylation of p70 S6 kinase was decreased following rapamycin treatment in vivo. Collectively these data demonstrate modulating p70 S6 kinase can be an effective antiviral strategy.


Subject(s)
Ribosomal Protein S6 Kinases, 70-kDa/drug effects , Rift Valley fever virus/drug effects , Signal Transduction/drug effects , Sirolimus/antagonists & inhibitors , Animals , Antiviral Agents/pharmacology , Apoptosis/drug effects , Cell Line , Chlorocebus aethiops , DNA Replication/drug effects , Disease Models, Animal , Eukaryotic Initiation Factor-4G/metabolism , Female , Immunohistochemistry , Liver/pathology , Mice , Mice, Inbred BALB C , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Rift Valley Fever/drug therapy , Rift Valley Fever/pathology , Rift Valley Fever/virology , Rift Valley fever virus/genetics , Rift Valley fever virus/growth & development , Rift Valley fever virus/pathogenicity , Sirolimus/metabolism , Sirolimus/therapeutic use , Survival Analysis , Vero Cells , Viral Load/drug effects , Virus Replication/drug effects
17.
PLoS One ; 9(10): e110873, 2014.
Article in English | MEDLINE | ID: mdl-25329596

ABSTRACT

This study aimed to characterize a soluble proteome of popliteal lymph nodes during lymphadenitis induced by intradermal injection of Bacillus anthracis Sterne spores in mice using tandem LC-MS/MS and reverse-phase protein microarray with antibodies specific to epitopes of phosphorylated proteins. More than 380 proteins were detected in the normal intra-nodal lymph, while the infectious process resulted in the profound changes in the protein abundances and appearance of 297 unique proteins. These proteins belong to an array of processes reflecting response to wounding, inflammation and perturbations of hemostasis, innate immune response, coagulation and fibrinolysis, regulation of body fluid levels and vascular disturbance among others. Comparison of lymph and serum revealed 83 common proteins. Also, using 71 antibodies specific to total and phosphorylated forms of proteins we carried initial characterization of circulating lymph phosphoproteome which brought additional information regarding signaling pathways operating in the lymphatics. The results demonstrate that the proteome of intra-nodal lymph serves as a sensitive sentinel of the processes occurring within the lymph nodes during infection. The acute innate response of the lymph nodes to anthrax is accompanied by cellular damage and inflammation with a large number of up- and down-regulated proteins many of which are distinct from those detected in serum. MS data are available via ProteomeXchange with identifier PXD001342.


Subject(s)
Anthrax/metabolism , Bacillus anthracis , Gene Expression Regulation , Lymph Nodes/metabolism , Proteome/biosynthesis , Skin Diseases, Bacterial/metabolism , Animals , Anthrax/pathology , Lymph Nodes/pathology , Mice , Skin Diseases, Bacterial/pathology
18.
Virology ; 468-470: 490-503, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25261871

ABSTRACT

New World alphaviruses belonging to the family Togaviridae are classified as emerging infectious agents and Category B select agents. Our study is focused on the role of the host extracellular signal-regulated kinase (ERK) in the infectious process of New World alphaviruses. Infection of human cells by Venezuelan equine encephalitis virus (VEEV) results in the activation of the ERK-signaling cascade. Inhibition of ERK1/2 by the small molecule inhibitor Ag-126 results in inhibition of viral multiplication. Ag-126-mediated inhibition of VEEV was due to potential effects on early and late stages of the infectious process. While expression of viral proteins was down-regulated in Ag-126 treated cells, we did not observe any influence of Ag-126 on the nuclear distribution of capsid. Finally, Ag-126 exerted a broad-spectrum inhibitory effect on New World alphavirus multiplication, thus indicating that the host kinase, ERK, is a broad-spectrum candidate for development of novel therapeutics against New World alphaviruses.


Subject(s)
Encephalitis Virus, Venezuelan Equine/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Signal Transduction/physiology , Virus Replication/physiology , Animals , Cell Line , Cell Survival , Encephalitis Virus, Eastern Equine/physiology , Encephalitis Virus, Western Equine/physiology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation, Viral , Phosphorylation , Tyrphostins/pharmacology , Virus Replication/drug effects
19.
Proteomics Clin Appl ; 8(11-12): 924-31, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25091122

ABSTRACT

PURPOSE: New treatment options for gastric cancer are in great demand. Histone deacetylases (HDACs) are exciting therapeutic targets, but only the class I HDACs 1, 2, and 3 have been studied in gastric cancer. We have investigated class IIa HDAC expression and inhibition in gastric cancer cells. EXPERIMENTAL DESIGN: We measured the level of 27 (phospho)proteins related to class IIa HDAC expression and function in ten laser-capture microdissection gastric tumor samples compared to patient-matched adjacent normal mucosa. Following, we evaluated class IIa HDAC inhibition by MC1568 in SNU-16 gastric cancer cells alone and in combination with cisplatin or docetaxel. RESULTS: We demonstrate for the first time an increase of HDAC4 in gastric tumor cells. HDAC4 inhibition had a synergistic effect with docetaxel treatment, shifting the cellular response from a cytostatic to a cytotoxic phenotype. This effect was associated with increased levels of cleaved caspases 3 and 9 and increased acetylated histone H3 Lys9/Lys14. CONCLUSIONS AND CLINICAL RELEVANCE: These data support in vivo studies investigating the potential clinical use of HDAC4 inhibitors in combination with docetaxel for the treatment of gastric cancer, lowering treatment doses of docetaxel to reduce the burden of adverse side effects on patients.


Subject(s)
Hydroxamic Acids/pharmacology , Pyrroles/pharmacology , Repressor Proteins/antagonists & inhibitors , Stomach Neoplasms/enzymology , Taxoids/pharmacology , Acetylation/drug effects , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Biomarkers/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Docetaxel , Drug Synergism , Female , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Repressor Proteins/metabolism , Stomach Neoplasms/pathology
20.
PLoS One ; 9(2): e86745, 2014.
Article in English | MEDLINE | ID: mdl-24586253

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) belongs to the genus Alphavirus, family Togaviridae. VEEV infection is characterized by extensive inflammation and studies from other laboratories implicated an involvement of the NF-κB cascade in the in vivo pathology. Initial studies indicated that at early time points of VEEV infection, the NF-κB complex was activated in cells infected with the TC-83 strain of VEEV. One upstream kinase that contributes to the phosphorylation of p65 is the IKKß component of the IKK complex. Our previous studies with Rift valley fever virus, which exhibited early activation of the NF-κB cascade in infected cells, had indicated that the IKKß component underwent macromolecular reorganization to form a novel low molecular weight form unique to infected cells. This prompted us to investigate if the IKK complex undergoes a comparable macromolecular reorganization in VEEV infection. Size-fractionated VEEV infected cell extracts indicated a macromolecular reorganization of IKKß in VEEV infected cells that resulted in formation of lower molecular weight complexes. Well-documented inhibitors of IKKß function, BAY-11-7082, BAY-11-7085 and IKK2 compound IV, were employed to determine whether IKKß function was required for the production of infectious progeny virus. A decrease in infectious viral particles and viral RNA copies was observed with inhibitor treatment in the attenuated and virulent strains of VEEV infection. In order to further validate the requirement of IKKß for VEEV replication, we over-expressed IKKß in cells and observed an increase in viral titers. In contrast, studies carried out using IKKß(-/-) cells demonstrated a decrease in VEEV replication. In vivo studies demonstrated that inhibitor treatment of TC-83 infected mice increased their survival. Finally, proteomics studies have revealed that IKKß may interact with the viral protein nsP3. In conclusion, our studies have revealed that the host IKKß protein may be critically involved in VEEV replication.


Subject(s)
Encephalitis Virus, Venezuelan Equine/physiology , Encephalomyelitis, Venezuelan Equine/metabolism , I-kappa B Kinase/metabolism , Animals , Cell Line , Down-Regulation/drug effects , Encephalomyelitis, Venezuelan Equine/pathology , Guinea Pigs , Host-Pathogen Interactions , Humans , Mice , NF-kappa B/metabolism , Neurons/virology , Nitriles/pharmacology , Signal Transduction/drug effects , Sulfones/pharmacology , Viral Load/drug effects , Viral Proteins/metabolism , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...