Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Mol Cell Proteomics ; 23(4): 100744, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38417630

ABSTRACT

NF-κB pathway is involved in inflammation; however, recent data shows its role also in cancer development and progression, including metastasis. To understand the role of NF-κB interactome dynamics in cancer, we study the complexity of breast cancer interactome in luminal A breast cancer model and its rearrangement associated with NF-κB modulation. Liquid chromatography-mass spectrometry measurement of 160 size-exclusion chromatography fractions identifies 5460 protein groups. Seven thousand five hundred sixty eight interactions among these proteins have been reconstructed by PrInCE algorithm, of which 2564 have been validated in independent datasets. NF-κB modulation leads to rearrangement of protein complexes involved in NF-κB signaling and immune response, cell cycle regulation, and DNA replication. Central NF-κB transcription regulator RELA co-elutes with interactors of NF-κB activator PRMT5, and these complexes are confirmed by AlphaPulldown prediction. A complementary immunoprecipitation experiment recapitulates RELA interactions with other NF-κB factors, associating NF-κB inhibition with lower binding of NF-κB activators to RELA. This study describes a network of pro-tumorigenic protein interactions and their rearrangement upon NF-κB inhibition with potential therapeutic implications in tumors with high NF-κB activity.


Subject(s)
Breast Neoplasms , NF-kappa B , Protein Interaction Maps , Transcription Factor RelA , Humans , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , NF-kappa B/metabolism , Transcription Factor RelA/metabolism , Protein Interaction Mapping , Signal Transduction , Cell Line, Tumor , Protein Binding , Protein-Arginine N-Methyltransferases/metabolism , Carcinogenesis/metabolism
2.
J Proteome Res ; 23(8): 2999-3011, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-38498986

ABSTRACT

Caspase-9 is traditionally considered the initiator caspase of the intrinsic apoptotic pathway. In the past decade, however, other functions beyond initiation/execution of cell death have been described including cell type-dependent regulation of proliferation, differentiation/maturation, mitochondrial, and endosomal/lysosomal homeostasis. As previous studies revealed nonapoptotic functions of caspases in osteogenesis and bone homeostasis, this study was performed to identify proteins and pathways deregulated by knockout of caspase-9 in mouse MC3T3-E1 osteoblasts. Data-independent acquisition-parallel accumulation serial fragmentation (diaPASEF) proteomics was used to compare protein profiles of control and caspase-9 knockout cells. A total of 7669 protein groups were quantified, and 283 upregulated/141 downregulated protein groups were associated with the caspase-9 knockout phenotype. The deregulated proteins were mainly enriched for those associated with cell migration and motility and DNA replication/repair. Altered migration was confirmed in MC3T3-E1 cells with the genetic and pharmacological inhibition of caspase-9. ABHD2, an established regulator of cell migration, was identified as a possible substrate of caspase-9. We conclude that caspase-9 acts as a modulator of osteoblastic MC3T3-E1 cell migration and, therefore, may be involved in bone remodeling and fracture repair.


Subject(s)
Caspase 9 , Cell Movement , Osteoblasts , Proteomics , Animals , Osteoblasts/metabolism , Osteoblasts/cytology , Mice , Proteomics/methods , Caspase 9/metabolism , Caspase 9/genetics , Cell Line , Gene Knockout Techniques
3.
Mol Cell Proteomics ; 21(2): 100188, 2022 02.
Article in English | MEDLINE | ID: mdl-34929376

ABSTRACT

Anterior gradient 2 (AGR2) is an endoplasmic reticulum (ER)-resident protein disulfide isomerase (PDI) known to be overexpressed in many human epithelial cancers and is involved in cell migration, cellular transformation, angiogenesis, and metastasis. This protein inhibits the activity of the tumor suppressor p53, and its expression levels can be used to predict cancer patient outcome. However, the precise network of AGR2-interacting partners and clients remains to be fully characterized. Herein, we used label-free quantification and also stable isotope labeling with amino acids in cell culture-based LC-MS/MS analyses to identify proteins interacting with AGR2. Functional annotation confirmed that AGR2 and its interaction partners are associated with processes in the ER that maintain intracellular metabolic homeostasis and participate in the unfolded protein response, including those associated with changes in cellular metabolism, energy, and redox states in response to ER stress. As a proof of concept, the interaction between AGR2 and PDIA3, another ER-resident PDI, was studied in more detail. Pathway analysis revealed that AGR2 and PDIA3 play roles in protein folding in ER, including post-translational modification and in cellular response to stress. We confirmed the AGR2-PDIA3 complex formation in cancer cells, which was enhanced in response to ER stress. Accordingly, molecular docking characterized potential quaternary structure of this complex; however, it remains to be elucidated whether AGR2 rather contributes to PDIA3 maturation in ER, the complex directly acts in cellular signaling, or mediates AGR2 secretion. Our study provides a comprehensive insight into the protein-protein interaction network of AGR2 by identifying functionally relevant proteins and related cellular and biochemical pathways associated with the role of AGR2 in cancer cells.


Subject(s)
Mucoproteins , Neoplasms , Oncogene Proteins , Protein Disulfide-Isomerases , Chromatography, Liquid , Humans , Molecular Docking Simulation , Mucoproteins/metabolism , Oncogene Proteins/metabolism , Protein Interaction Maps , Tandem Mass Spectrometry
4.
Proteomics ; 23(11): e2200408, 2023 06.
Article in English | MEDLINE | ID: mdl-36960851

ABSTRACT

Caspase-9 is the major apical caspase responsible for triggering the intrinsic apoptotic pathway. Our previous study indicated that specific inhibition of caspase-9 caused microscopically evident alterations in appearance of the primary chondrogenic cultures which cannot be explained by decrease in apoptosis. To describe a complex molecular background of this effect, proteomics analysis of control and caspase-9 inhibitor-treated chondrogenic cultures were performed. Proteins were extracted, identified and quantified using LC-MS in both data dependent and data independent acquisition (DIA) mode. While directDIA analysis of diaPASEF data obtained using timsTOF Pro LC-MS system revealed 7849 protein groups (Q-value <0.01), a parallel analysis of iTRAQ-2DLC-MS3 and conventional DIA-MS data identified only 5146 and 4098 protein groups, respectively, showing diaPASEF a superior method for the study. The detailed analysis of diaPASEF data disclosed 236/551 significantly down-/up-regulated protein groups after caspase-9 inhibition, respectively (|log2FC|>0.58, Q value <0.05). Classification of downregulated proteins revealed changes in extracellular matrix organization, collagen metabolism, and muscle system processes. Moreover, deregulations suggest a switch from glycolytic to lipid based metabolism in the inhibited cells. No essential changes were found in the proteins involved in apoptosis. The data indicate new non-apoptotic participation of caspases in chondrocyte homeostasis with potential applications in cartilage pathophysiology.


Subject(s)
Apoptosis , Chondrocytes , Caspase 9/metabolism , Caspase 9/pharmacology , Chondrocytes/metabolism , Signal Transduction , Caspases/metabolism , Caspases/pharmacology
5.
Cell Mol Biol Lett ; 28(1): 68, 2023 Aug 24.
Article in English | MEDLINE | ID: mdl-37620794

ABSTRACT

BACKGROUND: Desmocollin-1 (DSC1) is a desmosomal transmembrane glycoprotein that maintains cell-to-cell adhesion. DSC1 was previously associated with lymph node metastasis of luminal A breast tumors and was found to increase migration and invasion of MCF7 cells in vitro. Therefore, we focused on DSC1 role in cellular and molecular mechanisms in luminal A breast cancer and its possible therapeutic modulation. METHODS: Western blotting was used to select potential inhibitor decreasing DSC1 protein level in MCF7 cell line. Using atomic force microscopy we evaluated effect of DSC1 overexpression and modulation on cell morphology. The LC-MS/MS analysis of total proteome on Orbitrap Lumos and RNA-Seq analysis of total transcriptome on Illumina NextSeq 500 were performed to study the molecular mechanisms associated with DSC1. Pull-down analysis with LC-MS/MS detection was carried out to uncover DSC1 protein interactome in MCF7 cells. RESULTS: Analysis of DSC1 protein levels in response to selected inhibitors displays significant DSC1 downregulation (p-value ≤ 0.01) in MCF7 cells treated with NF-κB inhibitor parthenolide. Analysis of mechanic cell properties in response to DSC1 overexpression and parthenolide treatment using atomic force microscopy reveals that DSC1 overexpression reduces height of MCF7 cells and conversely, parthenolide decreases cell stiffness of MCF7 cells overexpressing DSC1. The LC-MS/MS total proteome analysis in data-independent acquisition mode shows a strong connection between DSC1 overexpression and increased levels of proteins LACRT and IGFBP5, increased expression of IGFBP5 is confirmed by RNA-Seq. Pathway analysis of proteomics data uncovers enrichment of proliferative MCM_BIOCARTA pathway including CDK2 and MCM2-7 after DSC1 overexpression. Parthenolide decreases expression of LACRT, IGFBP5 and MCM_BIOCARTA pathway specifically in DSC1 overexpressing cells. Pull-down assay identifies DSC1 interactions with cadherin family proteins including DSG2, CDH1, CDH3 and tyrosine kinase receptors HER2 and HER3; parthenolide modulates DSC1-HER3 interaction. CONCLUSIONS: Our systems biology data indicate that DSC1 is connected to mechanisms of cell cycle regulation in luminal A breast cancer cells, and can be effectively modulated by parthenolide.


Subject(s)
Desmocollins , Neoplasms , Chromatography, Liquid , Desmocollins/metabolism , Proteome , Tandem Mass Spectrometry , Humans , MCF-7 Cells , Sesquiterpenes/pharmacology
6.
Proteomics ; 22(7): e2100228, 2022 04.
Article in English | MEDLINE | ID: mdl-34902229

ABSTRACT

Renal cell carcinoma (RCC) represents 2.2% of all cancer incidences; however, prognostic or predictive RCC biomarkers at protein level are largely missing. To support proteomics research of localized and metastatic RCC, we introduce a new library of targeted mass spectrometry assays for accurate protein quantification in malignant and normal kidney tissue. Aliquots of 86 initially localized RCC, 75 metastatic RCC and 17 adjacent non-cancerous fresh frozen tissue lysates were trypsin digested, pooled, and fractionated using hydrophilic chromatography. The fractions were analyzed using LC-MS/MS on QExactive HF-X mass spectrometer in data-dependent acquisition (DDA) mode. A resulting spectral library contains 77,817 peptides representing 7960 protein groups (FDR = 1%). Further, we confirm applicability of this library on four RCC datasets measured in data-independent acquisition (DIA) mode, demonstrating a specific quantification of a substantially increased part of RCC proteome, depending on LC-MS/MS instrumentation. Impact of sample specificity of the library on the results of targeted DIA data extraction was demonstrated by parallel analyses of two datasets by two pan human libraries. The new RCC specific library has potential to contribute to better understanding the RCC development at molecular level, leading to new diagnostic and therapeutic targets.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Chromatography, Liquid , Humans , Proteome/metabolism , Tandem Mass Spectrometry
7.
Cancer Cell Int ; 22(1): 394, 2022 Dec 09.
Article in English | MEDLINE | ID: mdl-36494720

ABSTRACT

Cell migration and invasiveness significantly contribute to desirable physiological processes, such as wound healing or embryogenesis, as well as to serious pathological processes such as the spread of cancer cells to form tumor metastasis. The availability of appropriate methods for studying these processes is essential for understanding the molecular basis of cancer metastasis and for identifying suitable therapeutic targets for anti-metastatic treatment. This review summarizes the current status of these methods: In vitro methods for studying cell migration involve two-dimensional (2D) assays (wound-healing/scratch assay), and methods based on chemotaxis (the Dunn chamber). The analysis of both cell migration and invasiveness in vitro require more complex systems based on the Boyden chamber principle (Transwell migration/invasive test, xCELLigence system), or microfluidic devices with three-dimensional (3D) microscopy visualization. 3D culture techniques are rapidly becoming routine and involve multicellular spheroid invasion assays or array chip-based, spherical approaches, multi-layer/multi-zone culture, or organoid non-spherical models, including multi-organ microfluidic chips. The in vivo methods are mostly based on mice, allowing genetically engineered mice models and transplant models (syngeneic mice, cell line-derived xenografts and patient-derived xenografts including humanized mice models). These methods currently represent a solid basis for the state-of-the art research that is focused on understanding metastatic fundamentals as well as the development of targeted anti-metastatic therapies, and stratified treatment in oncology.

8.
Proteomics ; 20(7): e1900383, 2020 04.
Article in English | MEDLINE | ID: mdl-32061197

ABSTRACT

Transgelin is a protein reported to be a marker of several cancers. However, previous studies have shown both up- and down-regulation of transgelin in tumors when compared with non-tumor tissues and the mechanisms whereby transgelin may affect the development of cancer remain largely unknown. Transgelin is especially abundant in smooth muscle cells and is associated with actin stress fibers. These contractile structures participate in cell motility, adhesion, and the maintenance of cell morphology. Here, the role of transgelin in breast cancer is focused on. Initially, the effects of transgelin on cell migration of the breast cancer cell lines, BT 549 and PMC 42, is studied. Interestingly, transgelin silencing increased the migration of PMC 42 cells, but decreased the migration of BT 549 cells. To clarify these contradictory results, the changes in protein abundances after transgelin silencing in these two cell lines are analyzed using quantitative proteomics. The results confirmed the role of transgelin in the migration of BT 549 cells and suggest the involvement of transgelin in apoptosis and small molecule biochemistry in PMC 42 cells. The context-dependent function of transgelin reflects the different molecular backgrounds of these cell lines, which differ in karyotypes, mutation statuses, and proteome profiles.


Subject(s)
Apoptosis , Breast Neoplasms/genetics , Cell Movement , Gene Expression Regulation, Neoplastic , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/physiopathology , Cell Line, Tumor , Chromatography, Liquid , Down-Regulation , Female , Gene Knockdown Techniques , Gene Silencing , Humans , MCF-7 Cells , Microfilament Proteins/genetics , Muscle Proteins/genetics , Proteomics , Tandem Mass Spectrometry
9.
J Proteome Res ; 19(7): 2617-2630, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32343582

ABSTRACT

A specific form of endometrial cancer (EC) can develop in breast cancer patients previously treated with tamoxifen (ET), an antagonist of estrogen receptor (ER) that inhibits proliferation of ER positive breast cancer. ET tumors have a different phenotype than endometrial tumors, which typically develop de novo without previous exposure to tamoxifen (EN). Here we aimed to identify specific protein markers that could serve as specific molecular targets in either phenotype. A set of total 45 formalin-fixed paraffin-embedded (FFPE) endometrial tumor tissues and adjacent myometrium tissue samples were analyzed using LC-MS/MS in SWATH-MS mode. We found that calcyphosin (CAPS) levels were elevated in EN tumors compared to ET tumors. The higher CAPS level in EC tissue invading to myometrium supports its relationship to EC aggressiveness. Further, stathmin (STMN1) levels were found significantly elevated in ET versus EN tumors and significantly associated with patient survival. This finding connects elevated levels of this cell cycle regulating, proliferation-associated protein with tamoxifen exposure. In summary, using SWATH-MS we show that CAPS and STMN1 should be recognized as clinicopathologically different EC markers of which STMN1 is specifically connected with a previous tamoxifen exposition.


Subject(s)
Breast Neoplasms , Endometrial Neoplasms , Chromatography, Liquid , Endometrial Neoplasms/drug therapy , Female , Humans , Stathmin/genetics , Tamoxifen/adverse effects , Tandem Mass Spectrometry
10.
Proteomics ; 19(21-22): e1900073, 2019 11.
Article in English | MEDLINE | ID: mdl-31617665

ABSTRACT

Biological treatment of many cancers currently targets membrane bound receptors located on a cell surface. To identify novel membrane proteins associated with migration and metastasis of breast cancer cells, a more migrating subpopulation of MDA-MB-231 breast cancer cell line is selected and characterized. A high-resolution quantitative mass spectrometry with SILAC labeling is applied to analyze their surfaceome and it is compared with that of parental MDA-MB-231 cells. Among 824 identified proteins (FDR < 0.01), 128 differentially abundant cell surface proteins with at least one transmembrane domain are found. Of these, i) desmocollin-1 (DSC1) is validated as a protein connected with lymph node status of luminal A breast cancer, tumor grade, and Her-2 status by immunohistochemistry in the set of 96 primary breast tumors, and ii) catechol-O-methyltransferase is successfully verified as a protein associated with lymph node metastasis of triple negative breast cancer as well as with tumor grade by targeted data extraction from the SWATH-MS data of the same set of tissues. The findings indicate importance of both proteins for breast cancer development and metastasis and highlight the potential of biomarker validation strategy via targeted data extraction from SWATH-MS datasets.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catechol O-Methyltransferase/metabolism , Cell Movement , Desmocollins/metabolism , Lymphatic Metastasis/pathology , Proteomics , Breast Neoplasms/genetics , Catechol O-Methyltransferase/genetics , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement/genetics , Desmocollins/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Phenotype , Receptor, ErbB-2 , Survival Analysis , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Up-Regulation/genetics
11.
Proteomics ; 17(5)2017 03.
Article in English | MEDLINE | ID: mdl-27966270

ABSTRACT

Targeted mass spectrometry-based proteomics approaches enable the simultaneous and reproducible quantification of multiple protein analytes across numerous conditions in biology and clinical studies. These approaches involve e.g. selected reaction monitoring (SRM) typically conducted on a triple quadrupole mass spectrometer, its high-resolution variant named pseudo-SRM (p-SRM), carried out in a quadrupole coupled with an TOF analyzer (qTOF), and "sequential window acquisition of all theoretical spectra" (SWATH). Here we compared these methods in terms of signal-to-noise ratio (S/N), coefficient of variance (CV), fold change (FC), limit of detection and quantitation (LOD, LOQ). We have shown the highest S/N for p-SRM mode, followed by SRM and SWATH, demonstrating a trade-off between sensitivity and level of multiplexing for SRM, p-SRM, and SWATH. SRM was more sensitive than p-SRM based on determining their LOD and LOQ. Although SWATH has the worst S/N, it enables peptide multiplexing with post-acquisition definition of the targets, leading to better proteome coverage. FC between breast tumors of different clinical-pathological characteristics were highly correlated (R2 >0.97) across three methods and consistent with the previous study on 96 tumor tissues. Our technical note presented here, therefore, confirmed that outputs of all the three methods were biologically relevant and highly applicable to cancer research.


Subject(s)
Neoplasms/metabolism , Proteins/analysis , Proteomics/methods , Humans , Limit of Detection , Mass Spectrometry/instrumentation , Mass Spectrometry/methods , Neoplasms/chemistry , Signal-To-Noise Ratio
12.
Biochim Biophys Acta Proteins Proteom ; 1865(5): 488-498, 2017 May.
Article in English | MEDLINE | ID: mdl-28216224

ABSTRACT

Breast cancer is the most common and molecularly relatively well characterized malignant disease in women, however, its progression to metastatic cancer remains lethal for 78% of patients 5years after diagnosis. Novel markers could identify the high risk patients and their verification using quantitative methods is essential to overcome genetic, inter-tumor and intra-tumor variability and translate novel findings into cancer diagnosis and treatment. We recently identified 13 proteins associated with estrogen receptor, tumor grade and lymph node status, the key factors of breast cancer aggressiveness, using untargeted proteomics. Here we verified these findings in the same set of 96 tumors using targeted proteomics based on selected reaction monitoring with mTRAQ labeling (mTRAQ-SRM), transcriptomics and immunohistochemistry and validated in 5 independent sets of 715 patients using transcriptomics. We confirmed: (i) positive association of anterior gradient protein 2 homolog (AGR2) and periostin (POSTN) and negative association of annexin A1 (ANXA1) with estrogen receptor status; (ii) positive association of stathmin (STMN1), cofilin-1 (COF1), plasminogen activator inhibitor 1 RNA-binding protein (PAIRBP1) and negative associations of thrombospondin-2 (TSP2) and POSTN levels with tumor grade; and (iii) positive association of POSTN, alpha-actinin-4 (ACTN4) and STMN1 with lymph node status. This study highlights a panel of gene products that can contribute to breast cancer aggressiveness and metastasis, the understanding of which is important for development of more precise breast cancer treatment.


Subject(s)
Actin Depolymerizing Factors/biosynthesis , Breast Neoplasms/genetics , Cell Adhesion Molecules/biosynthesis , RNA-Binding Proteins/biosynthesis , Stathmin/biosynthesis , Thrombospondins/biosynthesis , Actin Depolymerizing Factors/genetics , Adult , Aged , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Cell Adhesion Molecules/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Prognosis , Proteomics , RNA-Binding Proteins/genetics , Receptors, Estrogen/genetics , Stathmin/genetics , Thrombospondins/genetics
13.
Cancer Invest ; 35(5): 313-324, 2017 May 28.
Article in English | MEDLINE | ID: mdl-28402678

ABSTRACT

Tamoxifen treatment in breast cancer patients is associated with increased risk of endometrial malignancies. Significantly, higher AGR2 expression was found in endometrial cancers that developed in women previously treated with tamoxifen compared to those who had not been exposed to tamoxifen. An association of elevated AGR2 level with myometrial invasion occurrence and invasion depth was also found. In vitro analyses identified a stimulatory effect of AGR2 on cellular proliferation. Although adverse tamoxifen effects on endometrial cells remain elusive, our work identifies elevated AGR2 as a candidate tamoxifen-dependent mechanism of action responsible for increased incidence of endometrial cancer.


Subject(s)
Adenocarcinoma/chemically induced , Adenocarcinoma/metabolism , Antineoplastic Agents, Hormonal/toxicity , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/chemically induced , Endometrial Neoplasms/chemically induced , Endometrium/drug effects , Proteins/metabolism , Tamoxifen/toxicity , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Endometrium/metabolism , Endometrium/pathology , Female , Humans , MCF-7 Cells , Mucoproteins , Neoplasm Invasiveness , Oncogene Proteins , Proteins/genetics , RNA Interference , Retrospective Studies , Risk Factors , Signal Transduction/drug effects , Transfection , Up-Regulation
14.
Exp Mol Pathol ; 102(2): 280-283, 2017 04.
Article in English | MEDLINE | ID: mdl-28238761

ABSTRACT

Expression of the AGR2 oncogene was shown to be associated with estrogen receptor positive tumors. This gene contributes to enhanced cellular proliferation, drug resistance, metastasis development and may also serve as a predictor of poor prognosis. However, our analysis of AGR2 expression in a subset of estrogen-receptor negative tumors revealed that AGR2 could also be upregulated in hormone-independent manner. AGR2 expression was shown to be significantly increased in HER2 positive breast tumors on both the mRNA and the protein level. Moreover, in a subset of estrogen- and progesterone-receptor negative and simultaneously HER2-positive cases, increased AGR2 expression significantly correlated with worse patient prognosis. Subsequent analysis of independent data sets either collected in our institute or obtained from Oncomine cancer microarray database confirmed all these findings.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Proteins/metabolism , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Cell Line, Tumor , Cell Proliferation , Female , Humans , Mucoproteins , Oncogene Proteins , Prognosis , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
15.
Mol Cell Proteomics ; 14(7): 1814-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25903579

ABSTRACT

Current prognostic factors are insufficient for precise risk-discrimination in breast cancer patients with low grade breast tumors, which, in disagreement with theoretical prognosis, occasionally form early lymph node metastasis. To identify markers for this group of patients, we employed iTRAQ-2DLC-MS/MS proteomics to 24 lymph node positive and 24 lymph node negative grade 1 luminal A primary breast tumors. Another group of 48 high-grade tumors (luminal B, triple negative, Her-2 subtypes) was also analyzed to investigate marker specificity for grade 1 luminal A tumors. From the total of 4405 proteins identified (FDR < 5%), the top 65 differentially expressed together with 30 previously identified and control markers were analyzed also at transcript level. Increased levels of carboxypeptidase B1 (CPB1), PDZ and LIM domain protein 2 (PDLIM2), and ring finger protein 25 (RNF25) were associated specifically with lymph node positive grade 1 tumors, whereas stathmin 1 (STMN1) and thymosin beta 10 (TMSB10) associated with aggressive tumor phenotype also in high grade tumors at both protein and transcript level. For CPB1, these differences were also observed by immunohistochemical analysis on tissue microarrays. Up-regulation of putative biomarkers in lymph node positive (versus negative) luminal A tumors was validated by gene expression analysis of an independent published data set (n = 343) for CPB1 (p = 0.00155), PDLIM2 (p = 0.02027) and RELA (p = 0.00015). Moreover, statistically significant connections with patient survival were identified in another public data set (n = 1678). Our findings indicate unique pro-metastatic mechanisms in grade 1 tumors that can include up-regulation of CPB1, activation of NF-κB pathway and changes in cell survival and cytoskeleton. These putative biomarkers have potential to identify the specific minor subpopulation of breast cancer patients with low grade tumors who are at higher than expected risk of recurrence and who would benefit from more intensive follow-up and may require more personalized therapy.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carboxypeptidase B/metabolism , Gene Expression Profiling/methods , NF-kappa B/metabolism , Proteomics/methods , Biomarkers, Tumor/genetics , Databases, Protein , Female , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Isotope Labeling , Kaplan-Meier Estimate , Neoplasm Grading , Neoplasm Metastasis , Reproducibility of Results
16.
Expert Rev Proteomics ; 12(6): 651-67, 2015.
Article in English | MEDLINE | ID: mdl-26456120

ABSTRACT

The concept of personalized medicine includes novel protein biomarkers that are expected to improve the early detection, diagnosis and therapy monitoring of malignant diseases. Tissues, biofluids, cell lines and xenograft models are the common sources of biomarker candidates that require verification of clinical value in independent patient cohorts. Targeted proteomics - based on selected reaction monitoring, or data extraction from data-independent acquisition based digital maps - now represents a promising mass spectrometry alternative to immunochemical methods. To date, it has been successfully used in a high number of studies answering clinical questions on solid malignancies: breast, colorectal, prostate, ovarian, endometrial, pancreatic, hepatocellular, lung, bladder and others. It plays an important role in functional proteomic experiments that include studying the role of post-translational modifications in cancer progression. This review summarizes verified biomarker candidates successfully quantified by targeted proteomics in this field and directs the readers who plan to design their own hypothesis-driven experiments to appropriate sources of methods and knowledge.


Subject(s)
Biomarkers, Tumor/chemistry , Mass Spectrometry/methods , Proteome/chemistry , Proteomics/methods , Animals , Biomarkers, Tumor/metabolism , Early Detection of Cancer/methods , Humans , Proteome/metabolism
17.
Proteomics ; 14(4-5): 426-40, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24272997

ABSTRACT

Metastases are responsible for most of the cases of death in patients with solid tumors. There is thus an urgent clinical need of better understanding the exact molecular mechanisms and finding novel therapeutics targets and biomarkers of metastatic disease of various tumors. Metastases are formed in a complicated biological process called metastatic cascade. Up to now, proteomics has enabled the identification of number of metastasis-associated proteins and potential biomarkers in cancer tissues, microdissected cells, model systems, and secretomes. Expression profiles and biological role of key proteins were confirmed in verification and functional experiments. This communication reviews these observations and analyses the methodological aspects of the proteomics approaches used. Moreover, it reviews contribution of current proteomics in the field of functional characterization and interactome analysis of proteins involved in various events in metastatic cascade. It is evident that ongoing technical progress will further increase proteome coverage and sample capacity of proteomics technologies, giving complex answers to clinical and functional questions asked.


Subject(s)
Biomarkers, Tumor/genetics , Neoplasm Metastasis/genetics , Neoplasms/genetics , Proteomics , Biomarkers, Tumor/metabolism , Humans , Microdissection , Neoplasm Metastasis/pathology , Neoplasms/metabolism , Neoplasms/pathology
18.
Expert Rev Proteomics ; 11(2): 149-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24476357

ABSTRACT

Transgelin is an abundant protein of smooth muscle cells, where its role has been primarily studied. As a protein affecting dynamics of the actin cytoskeleton via stabilization of actin filaments, transgelin is both directly and indirectly involved in many cancer-related processes such as migration, proliferation, differentiation or apoptosis. Transgelin was previously reviewed as a tumor suppressor; however, recent data based on a number of proteomics studies indicate its pro-tumorigenic role, for example, in colorectal or hepatocellular cancer. We summarize these contradictory observations in both clinical and functional proteomics projects and analyze the role of transgelin in tumors in detail. Generally, the expression and biological role of transgelin seem to differ among various types of tumor cells and stroma, and possibly change during tumor progression. We also overview the recent data on transgelin-2, a sequence homolog of transgelin, whose role in the tumor development might be contradictory to the role of transgelin.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Cytoskeletal Proteins/metabolism , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Neoplasms/metabolism , Actins/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis/pathology , Cell Movement , Cellular Senescence/physiology , Cytoskeletal Proteins/genetics , Epithelial-Mesenchymal Transition , Humans , Microfilament Proteins/genetics , Muscle Proteins/genetics , Muscle, Smooth/metabolism , Neoplasm Invasiveness/pathology , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Proteomics , Tumor Microenvironment
19.
Sci Data ; 11(1): 794, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39025866

ABSTRACT

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, and deeper proteome coverage is needed for its molecular characterization. We present comprehensive library of targeted mass spectrometry assays specific for TNBC and demonstrate its applicability. Proteins were extracted from 105 TNBC tissues and digested. Aliquots were pooled, fractionated using hydrophilic chromatography and analyzed by LC-MS/MS in data-dependent acquisition (DDA) parallel accumulation-serial fragmentation (PASEF) mode on timsTOF Pro LC-MS system. 16 individual lysates were analyzed in data-independent acquisition (DIA)-PASEF mode. Hybrid library was generated in Spectronaut software and covers 244,464 precursors, 168,006 peptides and 11,564 protein groups (FDR = 1%). Application of our library for pilot quantitative analysis of 16 tissues increased identification numbers in Spectronaut 18.5 and DIA-NN 1.8.1 software compared to library-free setting, with Spectronaut achieving the best results represented by 190,310 precursors, 140,566 peptides, and 10,463 protein groups. In conclusion, we introduce assay library that offers the deepest coverage of TNBC proteome to date. The TNBC library is available via PRIDE repository (PXD047793).


Subject(s)
Tandem Mass Spectrometry , Triple Negative Breast Neoplasms , Triple Negative Breast Neoplasms/genetics , Humans , Female , Chromatography, Liquid , Proteome , Proteomics/methods , Software
20.
Proteomics ; 13(7): 1053-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23303686

ABSTRACT

Proteomics profiling of intact proteins based on MALDI-TOF MS and derived platforms has been used in cancer biomarker discovery studies. This approach suffers from a number of limitations such as low resolution, low sensitivity, and that no knowledge is available on the identity of the respective proteins in the discovery mode. Nevertheless, it remains the most high-throughput, untargeted mode of clinical proteomics studies to date. Here we compare key protein separation and MS techniques available for protein biomarker identification in this type of studies and define reasons of uncertainty in protein peak identity. As a result of critical data analysis, we consider 3D protein separation and identification workflows as optimal procedures. Subsequently, we present a new protocol based on 3D LC-MS/MS with top-down at high resolution that enabled the identification of HNRNP A2/B1 intact peptide as correlating with the estrogen receptor expression in breast cancer tissues. Additional development of this general concept toward next generation, top-down based protein profiling at high resolution is discussed.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Neoplasm Proteins/isolation & purification , Neoplasm Proteins/metabolism , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Amino Acid Sequence , Female , Humans , Molecular Sequence Data , Neoplasm Proteins/chemistry , Peptides/chemistry , Peptides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL