Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters

Publication year range
1.
Analyst ; 148(5): 1041-1049, 2023 Feb 27.
Article in English | MEDLINE | ID: mdl-36723178

ABSTRACT

This work describes the development of a new approach to measure drug levels and lipid fingerprints in single living mammalian cells. Nanocapillary sampling is an approach that enables the selection and isolation of single living cells under microscope observation. Here, live single cell nanocapillary sampling is coupled to liquid chromatography for the first time. This allows molecular species to be separated prior to ionisation and improves measurement precision of drug analytes. The efficiency of transferring analytes from the sampling capillary into a vial was optimised in this work. The analysis was carried out using standard flow liquid chromatography coupled to widely available mass spectrometry instrumentation, highlighting opportunities for widespread adoption. The method was applied to 30 living cells, revealing cell-to-cell heterogeneity in the uptake of different drug molecules. Using this system, we detected 14-158 lipid features per single cell, revealing the association between bedaquiline uptake and lipid fingerprints.


Subject(s)
Lipids , Mammals , Animals , Mass Spectrometry/methods , Chromatography, Liquid/methods
2.
J Proteome Res ; 21(11): 2596-2608, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36264332

ABSTRACT

Lipids play a key role in many biological processes, and their accurate measurement is critical to unraveling the biology of diseases and human health. A high throughput HILIC-based (LC-MS) method for the semiquantitative screening of over 2000 lipids, based on over 4000 MRM transitions, was devised to produce an accessible and robust lipidomic screen for phospholipids in human plasma/serum. This methodology integrates many of the advantages of global lipid analysis with those of targeted approaches. Having used the method as an initial "wide class" screen, it can then be easily adapted for a more targeted analysis and quantification of key, dysregulated lipids. Robustness was assessed using 1550 continuous injections of plasma extracts onto a single column and via the evaluation of columns from 5 different batches of stationary phase. Initial screens in positive (239 lipids, 431 MRM transitions) and negative electrospray ionization (ESI) mode (232 lipids, 446 MRM transitions) were assessed for reproducibility, sensitivity, and dynamic range using analysis times of 8 min. The total number of lipids monitored using these screening methods was 433 with an overlap of 38 lipids in both modes. A polarity switching method for accurate quantification, using the same LC conditions, was assessed for intra- and interday reproducibility, accuracy, dynamic range, stability, carryover, dilution integrity, and matrix interferences and found to be acceptable. This polarity switching method was then applied to lipids important in the stratification of human prostate cancer samples.


Subject(s)
Lipidomics , Tandem Mass Spectrometry , Male , Humans , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Reproducibility of Results , Phospholipids
3.
Int J Mol Sci ; 23(9)2022 May 04.
Article in English | MEDLINE | ID: mdl-35563508

ABSTRACT

Post-traumatic OA (PTOA) is often triggered by injurious, high-impact loading events which result in rapid, excessive chondrocyte cell death and a phenotypic shift in residual cells toward a more catabolic state. As such, the identification of a disease-modifying OA drug (DMOAD) that can protect chondrocytes from death following impact injury, and thereby prevent cartilage degradation and progression to PTOA, would offer a novel intervention. We have previously shown that urocortin-1 (Ucn) is an essential endogenous pro-survival factor that protects chondrocytes from OA-associated pro-apoptotic stimuli. Here, using a drop tower PTOA-induction model, we demonstrate the extent of Ucn's chondroprotective role in cartilage explants exposed to excessive impact load. Using pathway-specific agonists and antagonists, we show that Ucn acts to block load-induced intracellular calcium accumulation through blockade of the non-selective cation channel Piezo1 rather than TRPV4. This protective effect is mediated primarily through the Ucn receptor CRF-R1 rather than CRF-R2. Crucially, we demonstrate that the chondroprotective effect of Ucn is maintained whether it is applied pre-impact or post-impact, highlighting the potential of Ucn as a novel DMOAD for the prevention of injurious impact overload-induced PTOA.


Subject(s)
Cartilage, Articular , Osteoarthritis , Cartilage, Articular/metabolism , Cell Death , Chondrocytes/metabolism , Humans , Ion Channels/metabolism , Osteoarthritis/etiology , Osteoarthritis/metabolism , Urocortins/metabolism , Urocortins/pharmacology
4.
J Proteome Res ; 20(3): 1705-1715, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33566619

ABSTRACT

The deployment of proteomic analysis in clinical studies represents a significant opportunity to detect and validate biomarkers in translational medicine, improve disease understanding, and provide baseline information on population health. However, comprehensive proteome studies usually employ nanoscale chromatography and often require several hours of analysis/sample. Here, we describe a high-throughput liquid chromatography tandem mass spectrometry (LC/MS/MS) methodology using 1 mm scale chromatography requiring only 15 min/sample, coupled to ion mobility-enabled mass spectrometry. The short run time effected a 6-fold increase in productivity compared with nanoscale LC/MS. The method demonstrated excellent reproducibility with retention time coefficient of variations of less than 0.05% and peak area reproducibility ranging from 5 to 15%. The 1 mm system produced similar chromatographic peak capacity values to the nanoscale miniaturized system, detecting 90% of the Escherichia coli proteins identified by the 75 µm LC/MS system (albeit based on only 75% of the peptides found by the latter). Application to the analysis of serum samples from a human prostate cancer study group resulted in the identification of a total of 533 proteins revealing the differential expression of proteins linked to patients receiving hormone-radiotherapy or undergoing surgery.


Subject(s)
Proteomics , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid , Chromatography, Liquid , Humans , Male , Reproducibility of Results
5.
Int J Mol Sci ; 22(9)2021 Apr 28.
Article in English | MEDLINE | ID: mdl-33925117

ABSTRACT

Taken with the growing importance of cathepsin-mediated substrate proteolysis in tumor biology and progression, the focus and emphasis placed on therapeutic design and development is coming into fruition. Underpinning this approach is the invariable progression from the direction of fully characterizing cathepsin protease members and their substrate targets, towards targeting such an interaction with tangible therapeutics. The two groups of such substrates that have gained much attention over the years are the pro- and anti- apoptotic protein intermediates from the extrinsic and intrinsic signaling arms of the apoptosis pathway. As proteins that are central to determining cellular fate, some of them present themselves as very favorable candidates for therapeutic targeting. However, considering that both anti- and pro- apoptotic signaling intermediates have been reported to be downstream substrates for certain activated cathepsin proteases, therapeutic targeting approaches based on greater selectivity do need to be given greater consideration. Herein, we review the relationships shared by the cathepsin proteases and the Bcl-2 homology domain proteins, in the context of how the topical approach of adopting 'BH3-mimetics' can be explored further in modulating the relationship between the anti- and pro- apoptotic signaling intermediates from the intrinsic apoptosis pathway and their upstream cathepsin protease regulators. Based on this, we highlight important future considerations for improved therapeutic design.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Cathepsins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Biomimetic Materials/pharmacology , Humans , Mitochondria/metabolism , Molecular Targeted Therapy , Peptide Fragments/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
6.
Int J Cancer ; 147(6): 1528-1534, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32010971

ABSTRACT

Cdc6 is a key replication licencing factor with a pivotal role in regulating the process of DNA replication, rendering it an important investigatory focus in tumourigenesis. Indeed, Cdc6 overexpression has been found to be a feature in certain tumours and has been associated as an early event in malignancies. With a focus on pancreatic cancer, there are evidence of its convergence in downstream pathways implicated in major genetic alterations found in pancreatic cancer, primarily KRAS. There is also data of its direct influence on protumourigenic processes as a transcriptional regulator, repressing the key tumour suppressor loci CDH1 (E-Cadherin) and influencing epithelial to mesenchymal transition (EMT). Moreover, gene amplification of Cdc6 as well as of E2F (an upstream regulator of Cdc6) have also been found to be a key feature in tumours overexpressing Cdc6, further highlighting this event as a potential driver of tumourigenesis. In this review, we summarise the evidence for the role of Cdc6 overexpression in cancer, specifically that of pancreatic cancer. More importantly, we recapitulate the role of Cdc6 as part of the DNA damage response and on senescence-an important antitumour barrier-in the context of pancreatic cancer. Finally, recent emerging observations suggest that the potential of the subcellular localisation of Cdc6 in inducing senescence. In this regard, we speculate and hypothesise potentially exploitable mechanisms in the context of inducing senescence via a novel pathway involving cytoplasmic retention of Cdc6 and Cyclin E.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinogenesis/genetics , Cell Cycle Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Nuclear Proteins/metabolism , Pancreatic Neoplasms/genetics , Antineoplastic Agents/therapeutic use , Carcinogenesis/drug effects , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Nucleus/metabolism , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cyclin E/metabolism , Cytoplasm/metabolism , DNA Damage/drug effects , DNA Damage/genetics , DNA Replication , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Amplification , Humans , Mutation , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Oncogene Proteins/metabolism , Pancreas/drug effects , Pancreas/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics
7.
Mol Cell Proteomics ; 16(3): 386-406, 2017 03.
Article in English | MEDLINE | ID: mdl-28062796

ABSTRACT

Approximately 800,000 leukemia and lymphoma cases are diagnosed worldwide each year. Burkitt's lymphoma (BL) and chronic lymphocytic leukemia (CLL) are examples of contrasting B-cell cancers; BL is a highly aggressive lymphoid tumor, frequently affecting children, whereas CLL typically presents as an indolent, slow-progressing leukemia affecting the elderly. The B-cell-specific overexpression of the myc and TCL1 oncogenes in mice induce spontaneous malignancies modeling BL and CLL, respectively. Quantitative mass spectrometry proteomics and isobaric labeling were employed to examine the biology underpinning contrasting Eµ-myc and Eµ-TCL1 B-cell tumors. Additionally, the plasma proteome was evaluated using subproteome enrichment to interrogate biomarker emergence and the systemic effects of tumor burden. Over 10,000 proteins were identified (q<0.01) of which 8270 cellular and 2095 plasma proteins were quantitatively profiled. A common B-cell tumor signature of 695 overexpressed proteins highlighted ribosome biogenesis, cell-cycle promotion and chromosome segregation. Eµ-myc tumors overexpressed several methylating enzymes and underexpressed many cytoskeletal components. Eµ-TCL1 tumors specifically overexpressed ER stress response proteins and signaling components in addition to both subunits of the interleukin-5 (IL5) receptor. IL5 treatment promoted Eµ-TCL1 tumor proliferation, suggesting an amplification of IL5-induced AKT signaling by TCL1. Tumor plasma contained a substantial tumor lysis signature, most prominent in Eµ-myc plasma, whereas Eµ-TCL1 plasma contained signatures of immune-response, inflammation and microenvironment interactions, with putative biomarkers in early-stage cancer. These findings provide a detailed characterization of contrasting B-cell tumor models, identifying common and specific tumor mechanisms. Integrated plasma proteomics allowed the dissection of a systemic response and a tumor lysis signature present in early- and late-stage cancers, respectively. Overall, this study suggests common B-cell cancer signatures exist and illustrates the potential of the further evaluation of B-cell cancer subtypes by integrative proteomics.


Subject(s)
Biomarkers, Tumor/analysis , Burkitt Lymphoma/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Proteomics/methods , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins/genetics , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Burkitt Lymphoma/genetics , Endoplasmic Reticulum Stress , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mass Spectrometry/methods , Mice , Mice, Transgenic
8.
Breast Cancer Res ; 20(1): 19, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29566726

ABSTRACT

BACKGROUND: Early-onset breast cancer (EOBC) affects about one in 300 women aged 40 years or younger and is associated with worse outcomes than later onset breast cancer. This study explored novel serum proteins as surrogate markers of prognosis in patients with EOBC. METHODS: Serum samples from EOBC patients (stages 1-3) were analysed using agnostic high-precision quantitative proteomics. Patients received anthracycline-based chemotherapy. The discovery cohort (n = 399) either had more than 5-year disease-free survival (DFS) (good outcome group, n = 203) or DFS of less than 2 years (poor outcome group, n = 196). Expressed proteins were assessed for differential expression between the two groups. Bioinformatics pathway and network analysis in combination with literature research were used to determine clinically relevant proteins. ELISA analysis against an independent sample set from the Prospective study of Outcomes in Sporadic versus Hereditary breast cancer (POSH) cohort (n = 181) was used to validate expression levels of the selected target. Linear and generalized linear modelling was applied to determine the effect of target markers, body mass index (BMI), lymph node involvement (LN), oestrogen receptor (ER), progesterone receptor and human epidermal growth factor receptor 2 status on patients' outcome. RESULTS: A total of 5346 unique proteins were analysed (peptide FDR p ≤ 0.05). Of these, 812 were differentially expressed in the good vs poor outcome groups and showed significant enrichment for the insulin signalling (p = 0.01) and the glycolysis/gluconeogenesis (p = 0.01) pathways. These proteins further correlated with interaction networks involving glucose and fatty acid metabolism. A consistent nodal protein to these metabolic networks was resistin (upregulated in the good outcome group, p = 0.009). ELISA validation demonstrated resistin to be upregulated in the good outcome group (p = 0.04), irrespective of BMI and ER status. LN involvement was the only covariate with a significant association with resistin measurements (p = 0.004). An ancillary in-silico observation was the induction of the inflammatory response, leucocyte infiltration, lymphocyte migration and recruitment of phagocytes (p < 0.0001, z-score > 2). Survival analysis showed that resistin overexpression was associated with improved DFS. CONCLUSIONS: Higher circulating resistin correlated with node-negative patients and longer DFS independent of BMI and ER status in women with EOBC. Overexpression of serum resistin in EOBC may be a surrogate indicator of improved prognosis.


Subject(s)
Blood Proteins/genetics , Breast Neoplasms/blood , Proteomics , Resistin/blood , Adult , Biomarkers, Tumor/blood , Body Mass Index , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Insulin Resistance , Lymph Nodes/pathology , Neoplastic Cells, Circulating/pathology , Prognosis , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics
9.
Surgeon ; 13(5): 271-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25736469

ABSTRACT

BACKGROUND: Breast cancer is a heterogeneous disease. Yet, many molecular players and mechanisms behind the complexity of its clinical behaviour remain unknown, and advances in biomedical research are expected to unravel novel molecular discoveries in breast and other cancers. Clinical proteomics is currently experiencing rapid advances in technology that promise new means to improve breast cancer early diagnosis, stratification, and treatment response. METHODS: We reviewed recent literature adopting clinical proteomics in breast cancer research. FINDINGS: This review highlights the principles, advantages, limitations, discoveries and future prospects of recent clinical proteomics discovery efforts in breast cancer research. CONCLUSION: Numerous proteomic studies of breast cancer have been accomplished aiming to aid the development of personalised therapies, increase understanding of post treatment relapse, and help improve prediction of patient prognosis. This has led to the possible identification of profiles refining breast cancer subtypes and the discovery of novel biomarkers pointing towards diagnostic and prognostic potential.


Subject(s)
Biomarkers, Tumor/metabolism , Biomedical Research/methods , Breast Neoplasms/diagnosis , Proteomics/methods , Breast Neoplasms/metabolism , Female , Humans , Prognosis
10.
J Proteome Res ; 13(11): 5094-105, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25072778

ABSTRACT

Linking gender-specific differences to the molecular etiology of obesity has been largely based on genomic and transcriptomic evidence lacking endophenotypic insight and is not applicable to the extracellular fluid compartments, or the milieu intérieur, of the human body. To address this need, this study profiled the whole serum proteomes of age-matched nondiabetic overweight and obese females (n = 28) and males (n = 31) using a multiplex design with pooled biological and technical replicates. To bypass basic limitations of immunodepletion-based strategies, subproteome enrichment by size-exclusion chromatography (SuPrE-SEC) followed by iTRAQ 2D-LC-nESI-FTMS analysis was used. The study resulted in the reproducible analysis of 2472 proteins (peptide FDR < 5%, q < 0.05). A total of 248 proteins exhibited significant modulation between men and women (p < 0.05) that mapped to pathways associated with ß-estradiol, lipid and prostanoid metabolism, vitamin D function, immunity/inflammation, and the complement and coagulation cascades. This novel endophenotypic signature of gender-specific differences in whole serum confirmed and expanded the results of previous physiologic and pharmacologic studies exploring sexual dimorphism at the genomic and transcriptomic level in tissues and cells. Conclusively, the multifactorial and pleiotropic nature of human obesity exhibits sexual dimorphism in the circulating proteome of importance to clinical study design.


Subject(s)
Blood Proteins/metabolism , Chromatography, Gel/methods , Obesity/blood , Overweight/blood , Proteomics/methods , Spectrometry, Mass, Electrospray Ionization/methods , Adult , Blood Proteins/analysis , Blood Proteins/genetics , Female , Humans , Male , Middle Aged , Obesity/metabolism , Overweight/metabolism , Protein Interaction Maps , Sex Characteristics , Sex Factors , Transcriptome
12.
Nutrients ; 16(3)2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38337692

ABSTRACT

We previously demonstrated a beneficial effect of high-dose vitamin D in pregnancy on offspring bone and dental health. Here, we investigated the effect of maternal dietary patterns during pregnancy on the risk of bone fractures, bone mineralization and enamel defects until age 6 years in the offspring. Further, the influence of diet on the effect of high-dose vitamin D was analyzed in the COPSAC2010 mother-child cohort including 623 mother-child pairs. A weighted network analysis on FFQs revealed three specific maternal dietary patterns that associated (Bonferroni p < 0.05) with both offspring bone and dental health. The effect of prenatal high-dose (2800 IU/day) vs. standard-dose (400 IU/day) vitamin D on offspring bone mineral content (adjusted mean difference (aMD): 33.29 g, 95% CI: 14.48-52.09, p < 0.001), bone mineral density (aMD: 0.02 g/cm2 (0.01-0.04), p < 0.001), fracture risk (adjusted incidence rate ratio: 0.36 (0.16-0.84), p = 0.02), and enamel defects in primary (adjusted odds ratio (aOR): 0.13 (0.03-0.58), p < 0.01) and permanent molars (aOR: 0.25; (0.10-0.63), p < 0.01) was most pronounced when mothers had lower intake of fruit, vegetables, meat, eggs, sweets, whole grain, offal and fish. This study suggests that prenatal dietary patterns influence offspring bone and dental development, and should be considered in order to obtain the full benefits of vitamin D to enhance personalized supplementation strategy.


Subject(s)
Fractures, Bone , Vitamin D , Pregnancy , Female , Animals , Humans , Child , Calcification, Physiologic , Diet , Vitamins/pharmacology , Fractures, Bone/epidemiology , Fractures, Bone/etiology , Fractures, Bone/prevention & control , Bone Density , Dietary Supplements , Dental Enamel
13.
J Mol Cell Cardiol ; 59: 139-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23510923

ABSTRACT

Cardiac hypertrophy involves the growth of heart muscle cells and is driven by faster protein synthesis which involves increased ribosome biogenesis. However, the signaling pathways that link hypertrophic stimuli to faster ribosome production remain to be identified. Here we have investigated the signaling pathways which promote ribosomal RNA synthesis in cardiomyocytes in response to hypertrophic stimulation. We employed a new non-radioactive labeling approach and show that the hypertrophic agent phenylephrine (PE) stimulates synthesis of 18S rRNA (made by RNA polymerase I) and 5S rRNA (produced by RNA polymerase III) in adult cardiomyocytes. In many settings, rRNA synthesis is driven by rapamycin-sensitive signaling through mammalian target of rapamycin complex 1 (mTORC1). However, the activation of rRNA synthesis by PE is not inhibited by rapamycin, indicating that its regulation involves other signaling pathways. PE stimulates MEK/ERK signaling in these cells. Inhibition of this pathway blocks the ability of PE to activate synthesis of 18S and 5S rRNA. Furthermore, BI-D1870, an inhibitor of the p90(RSK)s, protein kinases which are activated by ERK, blocks PE-activated rRNA synthesis, as did a second p90(RSK) inhibitor, SL0101. BI-D1870 also inhibits the PE-stimulated association of RNA polymerase I with the rRNA promoter. These findings show that signaling via MEK/ERK/p90(RSK), not mTORC1, drives rRNA synthesis in adult cardiomyocytes undergoing hypertrophy. This is important both for our understanding of the mechanisms that control ribosome production and, potentially, for the management of cardiac hypertrophy.


Subject(s)
Cardiomegaly/chemically induced , Cardiomegaly/etiology , Myocytes, Cardiac/metabolism , Phenylephrine/pharmacology , RNA, Ribosomal/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Animals , Chromatin Immunoprecipitation , Male , Myocytes, Cardiac/drug effects , Rats , Rats, Sprague-Dawley , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
14.
J Proteome Res ; 12(5): 2078-89, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23510160

ABSTRACT

The ThinPrep cervical smear is widely used in clinical practice for the cytological and molecular screening against abnormal cells and Human Papillomavirus (HPV) infection. Current advancements made to LC-MS proteomics include the use of stable isotope labeling for the in-depth analysis of proteins in complex clinical specimens. Such approaches have yet to be realized for ThinPrep clinical specimens. In this study, an LC-MS method based on isobaric (iTRAQ) labeling and high-resolution FT-Orbitrap mass spectrometry was used for the proteomic analysis of 23 human ThinPrep smear specimens. Tandem mass spectrometry analysis was performed with both nitrogen high collision dissociation (HCD MS/MS) and helium collision induced dissociation (CID MS/MS) peptide fragmentation modes. The analysis of three 8-plex sample sets yielded the identification of over 3200 unique proteins at FDR < 1%, of which over 2300 proteins were quantitatively profiled in at least one of the three experiments. The interindividual variability served to define the required sample size needed to identify significant protein expression differences. The degree of in-depth proteome coverage allowed the detection of 6 HPV-derived proteins including the high-risk HPV16 type in the specimens tested. The presence of the HPV strains of origin was also confirmed with PCR-hybridization molecular methods. This proof-of-principle study constitutes the first ever report on the nontargeted analysis of HPV proteins in human ThinPrep clinical specimens with high-resolution mass spectrometry. A further testament to the sensitivity and selectivity of the proposed study method was the confident detection of a significant number of phosphopeptides in these specimens.


Subject(s)
Human papillomavirus 16/metabolism , Papillomavirus Infections/metabolism , Proteome/metabolism , Uterine Cervical Neoplasms/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Female , Humans , Isotope Labeling , Molecular Sequence Annotation , Molecular Sequence Data , Papillomavirus Infections/diagnosis , Proteome/chemistry , Proteomics , Sensitivity and Specificity , Tandem Mass Spectrometry/methods , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/virology , Vaginal Smears , Viral Proteins/chemistry
15.
J Proteome Res ; 12(4): 1591-603, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23418717

ABSTRACT

A naturally occurring benzofuran derivative, Ebenfuran III (Eb III), was investigated for its antiproliferative effects using the DU-145 prostate cell line. Eb III was isolated from Onobrychis ebenoides of the Leguminosae family, a plant endemic in Central and Southern Greece. We have previously reported that Eb III exerts significant cytotoxic effects on certain cancer cell lines. This effect is thought to occur via the isoprenyl moiety at the C-5 position of the molecule. The study aim was to gain a deeper understanding of the pharmacological effect of Eb III on DU-145 cell death at the translational level using a relative quantitative and temporal proteomics approach. Proteins extracted from the cell pellets were subjected to solution phase trypsin proteolysis followed by iTRAQ-labeling. The labeled tryptic peptide extracts were then fractionated using strong cation exchange chromatography and the fractions were analyzed by nanoflow reverse phase ultraperformance liquid chromatography-nanoelectrospray ionization-tandem mass spectrometry analysis using a hybrid QqTOF platform. Using this approach, we compared the expression levels of 1360 proteins analyzed at ≤ 1% global protein false discovery rate (FDR), commonly present in untreated (control, vehicle only) and Eb III-treated cells at the different exposure time points. Through the iterative use of Ingenuity Pathway Analysis with hierarchical clustering of protein expression patterns, followed by bibliographic research, the temporal regulation of the Calpain-1, ERK2, PAR-4, RAB-7, and Bap31 proteins were identified as potential nodes of multipathway convergence to Eb III induced DU-145 cell death. These proteins were further verified with Western blot analysis. This gel-free, quantitative 2DLC-MS/MS proteomics method effectively captured novel modulated proteins in the DU-145 cell line as a response to Eb III treatment. This approach also provided greater insight to the multifocal and combinatorial signaling pathways implicated in Eb III-induced cell death.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Benzofurans/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Proteins/metabolism , Resorcinols/pharmacology , Apoptosis Regulatory Proteins/metabolism , Calpain/metabolism , Cell Death/drug effects , Cell Line, Tumor , Chromatography, Reverse-Phase/methods , Cluster Analysis , Humans , Male , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Prostatic Neoplasms/pathology , Proteins/analysis , Tandem Mass Spectrometry/methods , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
16.
J Am Chem Soc ; 135(28): 10418-25, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23796364

ABSTRACT

Hypoxia inducible factor-1 (HIF-1) is a heterodimeric transcription factor that acts as the master regulator of cellular response to reduced oxygen levels, thus playing a key role in the adaptation, survival, and progression of tumors. Here we report cyclo-CLLFVY, identified from a library of 3.2 million cyclic hexapeptides using a genetically encoded high-throughput screening platform, as an inhibitor of the HIF-1α/HIF-1ß protein-protein interaction in vitro and in cells. The identified compound inhibits HIF-1 dimerization and transcription activity by binding to the PAS-B domain of HIF-1α, reducing HIF-1-mediated hypoxia response signaling in a variety of cell lines, without affecting the function of the closely related HIF-2 isoform. The reported cyclic peptide demonstrates the utility of our high-throughput screening platform for the identification of protein-protein interaction inhibitors, and forms the starting point for the development of HIF-1 targeted cancer therapeutics.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia , Peptides, Cyclic/pharmacology , Signal Transduction/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , MCF-7 Cells , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Structure-Activity Relationship
17.
Expert Rev Proteomics ; 10(4): 337-54, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23992417

ABSTRACT

Despite intense global efforts, no new clinical and/or viable biomarkers have been established to overcome the limitation of the prostate specific antigen in the early diagnosis and prognosis of prostate cancer (PCa). The current proteomic approaches to PCa biomarker discovery, each have distinct advantages and disadvantages, yet when combined hold real promise in the coming years. One key approach to this effort is the development of non-targeted, depletion-free and quantitative liquid chromatography-ultra high resolution tandem mass spectrometry (LC-MS) pipelines for the systems-wide interrogation of the diverse proteomes encompassed in whole tissue and blood serum or plasma. Derived quantitative proteomes can be decoded for their biomedical relevance with advanced bioinformatics and bibliographic mining to yield promising 'molecular portraits' that can gauge prostatic disease at the serological level. Their functional annotation, although potentially useful, is beyond our current level of biological understanding and should not be requisite for their effective use in the clinical monitoring of prostatic disease.


Subject(s)
Biomarkers, Tumor/metabolism , Early Detection of Cancer/methods , Prostatic Neoplasms/diagnosis , Early Detection of Cancer/trends , Humans , Male , Medical Oncology/methods , Medical Oncology/trends , Prostatic Neoplasms/metabolism , Proteome/metabolism , Proteomics/methods , Proteomics/trends
18.
Am J Pathol ; 180(3): 929-939, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22210479

ABSTRACT

Cardiac remodeling and hypertrophy are the pathological consequences of cardiovascular disease and are correlated with its associated mortality. Activity of the transcription factor NF-κB is increased in the diseased heart; however, our present understanding of how the individual subunits contribute to cardiovascular disease is limited. We assign a new role for the c-Rel subunit as a stimulator of cardiac hypertrophy and fibrosis. We discovered that c-Rel-deficient mice have smaller hearts at birth, as well as during adulthood, and are protected from developing cardiac hypertrophy and fibrosis after chronic angiotensin infusion. Results of both gene expression and cross-linked chromatin immunoprecipitation assay analyses identified transcriptional activators of hypertrophy, myocyte enhancer family, Gata4, and Tbx proteins as Rel gene targets. We suggest that the p50 subunit could limit the prohypertrophic actions of c-Rel in the normal heart, because p50 overexpression in H9c2 cells repressed c-Rel levels and the absence of cardiac p50 was associated with increases in both c-Rel levels and cardiac hypertrophy. We report for the first time that c-Rel is highly expressed and confined to the nuclei of diseased adult human hearts but is restricted to the cytoplasm of normal cardiac tissues. We conclude that c-Rel-dependent signaling is critical for both cardiac remodeling and hypertrophy. Targeting its activities could offer a novel therapeutic strategy to limit the effects of cardiac disease.


Subject(s)
Cardiomegaly/etiology , Myocardium/pathology , NF-kappa B/physiology , Proto-Oncogene Proteins c-rel/physiology , Angiotensins/pharmacology , Animals , Blood Pressure/physiology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Fibrosis , Gene Deletion , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B p50 Subunit/metabolism , NF-kappa B p50 Subunit/physiology , Proto-Oncogene Proteins c-rel/deficiency , Proto-Oncogene Proteins c-rel/genetics , Signal Transduction/physiology , Ventricular Remodeling/physiology
19.
J Nanobiotechnology ; 11: 20, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23809777

ABSTRACT

BACKGROUND: In ultrasonic micro-devices, contrast agent micro-bubbles are known to initiate cavitation and streaming local to cells, potentially compromising cell viability. Here we investigate the effects of US alone by omitting contrast agent and monitoring cell viability under moderate-to-extreme ultrasound-related stimuli. RESULTS: Suspended H9c2 cardiac myoblasts were exposed to ultrasonic fields within a glass micro-capillary and their viability monitored under different US-related stimuli. An optimal injection flow rate of 2.6 mL/h was identified in which, high viability was maintained (~95%) and no mechanical stress towards cells was evident. This flow rate also allowed sufficient exposure of cells to US in order to induce bioeffects (~5 sec), whilst providing economical sample collection and processing times. Although the transducer temperature increased from ambient 23°C to 54°C at the maximum experimental voltage (29 Vpp), computational fluid dynamic simulations and controls (absence of US) revealed that the cell medium temperature did not exceed 34°C in the pressure nodal plane. Cells exposed to US amplitudes ranging from 0-29 Vpp, at a fixed frequency sweep period (tsw = 0.05 sec), revealed that viability was minimally affected up to ~15 Vpp. There was a ~17% reduction in viability at 21 Vpp, corresponding to the onset of Rayleigh-like streaming and a ~60% reduction at 29 Vpp, corresponding to increased streaming velocity or the potential onset of cavitation. At a fixed amplitude (29 Vpp) but with varying frequency sweep period (tsw = 0.02-0.50 sec), cell viability remained relatively constant at tsw ≥ 0.08 sec, whilst viability reduced at tsw < 0.08 sec and minimum viability recorded at tsw = 0.05 sec. CONCLUSION: The absence of CA has enabled us to investigate the effect of US alone on cell viability. Moderate-to-extreme US-related stimuli of cells have allowed us to discriminate between stimuli that maintain high viability and stimuli that significantly reduce cell viability. Results from this study may be of potential interest to researchers in the field of US-induced intracellular drug delivery and ultrasonic manipulation of biological cells.


Subject(s)
Microfluidics/methods , Myocytes, Cardiac/cytology , Ultrasonics , Animals , Cell Survival , Oscillometry , Rats , Rheology , Temperature
20.
Cancers (Basel) ; 15(4)2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36831393

ABSTRACT

Prostate cancer is the most common malignant tumour in men. Improved testing for diagnosis, risk prediction, and response to treatment would improve care. Here, we identified a proteomic signature of prostate cancer in peripheral blood using data-independent acquisition mass spectrometry combined with machine learning. A highly predictive signature was derived, which was associated with relevant pathways, including the coagulation, complement, and clotting cascades, as well as plasma lipoprotein particle remodeling. We further validated the identified biomarkers against a second cohort, identifying a panel of five key markers (GP5, SERPINA5, ECM1, IGHG1, and THBS1) which retained most of the diagnostic power of the overall dataset, achieving an AUC of 0.91. Taken together, this study provides a proteomic signature complementary to PSA for the diagnosis of patients with localised prostate cancer, with the further potential for assessing risk of future development of prostate cancer. Data are available via ProteomeXchange with identifier PXD025484.

SELECTION OF CITATIONS
SEARCH DETAIL