Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Br J Haematol ; 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38651345

ABSTRACT

Despite advancements in utilizing genetic markers to enhance acute myeloid leukaemia (AML) outcome prediction, significant disease heterogeneity persists, hindering clinical management. To refine survival predictions, we assessed the transcriptome of non-acute promyelocytic leukaemia chemotherapy-treated AML patients from five cohorts (n = 975). This led to the identification of a 4-gene prognostic index (4-PI) comprising CYP2E1, DHCR7, IL2RA and SQLE. The 4-PI effectively stratified patients into risk categories, with the high 4-PI group exhibiting TP53 mutations and cholesterol biosynthesis signatures. Single-cell RNA sequencing revealed enrichment for leukaemia stem cell signatures in high 4-PI cells. Validation across three cohorts (n = 671), including one with childhood AML, demonstrated the reproducibility and clinical utility of the 4-PI, even using cost-effective techniques like real-time quantitative polymerase chain reaction. Comparative analysis with 56 established prognostic indexes revealed the superior performance of the 4-PI, highlighting its potential to enhance AML risk stratification. Finally, the 4-PI demonstrated to be potential marker to reclassified patients from the intermediate ELN2017 category to the adverse category. In conclusion, the 4-PI emerges as a robust and straightforward prognostic tool to improve survival prediction in AML patients.

2.
J Ovarian Res ; 16(1): 232, 2023 Nov 29.
Article in English | MEDLINE | ID: mdl-38031074

ABSTRACT

BACKGROUND: The epithelial-mesenchymal transition (EMT) promotes cell signaling and morphology alterations, contributing to cancer progression. Exosomes, extracellular vesicles containing proteins involved in cell-cell communication, have emerged as a potential source of biomarkers for several diseases. METHODS: Our aim was to assess the proteome content of exosomes secreted after EMT-induction to identify potential biomarkers for ovarian cancer classification. EMT was induced in the ovarian cancer cell line CAOV3 by treating it with EGF (10 ng/mL) for 96 h following 24 h of serum deprivation. Subsequently, exosomes were isolated from the supernatant using selective centrifugation after decellularization, and their characteristics were determined. The proteins present in the exosomes were extracted, identified, and quantified using Label-Free-Quantification (LFQ) via Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS). To identify potential biomarkers, the obtained proteomic data was integrated with the TGGA database for mRNA expression using principal component analysis and a conditional inference tree. RESULTS: The exosomes derived from CAOV3 cells exhibited similar diameter and morphology, measuring approximately 150 nm, regardless of whether they were subjected to EMT stimulation or not. The proteomic analysis of proteins from CAOV3-derived exosomes revealed significant differential regulation of 157 proteins, with 100 showing upregulation and 57 downregulation upon EMT induction. Further comparison of the upregulated proteins with the TCGA transcriptomic data identified PLAU, LAMB1, COL6A1, and TGFB1 as potential biomarkers of the mesenchymal HGSOC subtype. CONCLUSIONS: The induction of EMT, the isolation of exosomes, and the subsequent proteomic analysis highlight potential biomarkers for an aggressive ovarian cancer subtype. Further investigation into the role of these proteins is warranted to enhance our understanding of ovarian cancer outcomes.


Subject(s)
Exosomes , Ovarian Neoplasms , Female , Humans , Exosomes/metabolism , Epithelial-Mesenchymal Transition/genetics , Proteomics , Chromatography, Liquid , Tandem Mass Spectrometry , Biomarkers/metabolism , Ovarian Neoplasms/metabolism , Cell Line, Tumor
3.
Mol Cell Proteomics ; 20: 100091, 2021.
Article in English | MEDLINE | ID: mdl-33971369

ABSTRACT

Non-T cell activation linker (NTAL) membrane protein depletion from lipid rafts by alkylphospholipids or downregulation by shRNA knockdown decreases cell viability through regulation of the Akt/PI3K pathway in mantle cell lymphoma and acute promyelocytic leukemia cells. Here, we confirmed that the knockdown of NTAL in acute myeloid leukemia (AML) cell lines was associated with decreased cell proliferation and survival. Similarly, a xenograft model using AML cells transduced with NTAL-shRNA and transplanted into immunodeficient mice led to a 1.8-fold decrease in tumor burden. Using immunoprecipitation, LC-MS/MS analysis, and label-free protein quantification, we identified interactors of NTAL in two AML cell lines. By evaluating the gene expression signatures of the NTAL protein interactors using the PREdiction of Clinical Outcomes from Genomic Profiles database, we found that 12 NTAL interactors could predict overall survival in AML, in at least two independent cohorts. In addition, patients with AML exhibiting a high expression of NTAL and its interactors were associated with a leukemic granulocyte-macrophage progenitor-like state. Taken together, our data provide evidence that NTAL and its protein interactors are relevant to AML cell proliferation and survival and represent potential therapeutic targets for granulocyte-macrophage progenitor-like leukemias.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Leukemia, Myeloid, Acute/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival , Humans , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Protein Interaction Maps , Proto-Oncogene Proteins c-akt/metabolism , Survival Analysis , Transcriptome
4.
Int J Mol Sci ; 21(13)2020 07 01.
Article in English | MEDLINE | ID: mdl-32630308

ABSTRACT

Gliomas are responsible for more than 60% of all primary brain tumors. Glioblastoma multiforme (GBM), a grade IV tumor (WHO), is one of the most frequent and malignant gliomas. Despite two decades of advances in the discovery of new markers for GBM, the chemotherapy of choice falls to temozolomide after surgery and radiotherapy, which are not enough to increase the survival of patients to more than 15 months. It is urgent to discover new anti-glioma compounds. Many compounds derived from natural products have been used in the development of anti-tumor drugs. In this work, we have screened six low molecular weight sesquiterpene lactones, isolated from Eremanthus spp., and studied their function as anti-proliferative agents against GBM strains. We demonstrated that two of them, goyazensolide and lychnofolide, were effective in reducing cell viability, preventing the formation of anchorage-dependent colony and were able to pass through a mimetic blood-brain barrier making them candidates for glioma therapy, being more potent than temozolomide, according to in vitro assays for the cell lines tested. Proteomic analysis revealed a number of altered proteins involved in glycolytic metabolism and cellular catabolism.


Subject(s)
Lactones/pharmacology , Vernonia/metabolism , Antineoplastic Agents/pharmacology , Asteraceae , Blood-Brain Barrier/metabolism , Brain Neoplasms/metabolism , Brazil , Bridged-Ring Compounds/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Furans/pharmacology , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioma/metabolism , Humans , Lactones/metabolism , Plant Extracts/pharmacology , Sesquiterpenes/pharmacology , Sesterterpenes/pharmacology , Vernonia/physiology
5.
Sci Rep ; 10(1): 10315, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32587277

ABSTRACT

Non-T cell activation linker (NTAL) is a lipid raft-membrane protein expressed by normal and leukemic cells and involved in cell signaling. In acute promyelocytic leukemia (APL), NTAL depletion from lipid rafts decreases cell viability through regulation of the Akt/PI3K pathway. The role of NTAL in APL cell processes, and its association with clinical outcome, has not, however, been established. Here, we show that reduced levels of NTAL were associated with increased all-trans retinoic acid (ATRA)-induced differentiation, generation of reactive oxygen species, and mitochondrial dysfunction. Additionally, NTAL-knockdown (NTAL-KD) in APL cell lines led to activation of Ras, inhibition of Akt/mTOR pathways, and increased expression of autophagy markers, leading to an increased apoptosis rate following arsenic trioxide treatment. Furthermore, NTAL-KD in NB4 cells decreased the tumor burden in (NOD scid gamma) NSG mice, suggesting its implication in tumor growth. A retrospective analysis of NTAL expression in a cohort of patients treated with ATRA and anthracyclines, revealed that NTAL overexpression was associated with a high leukocyte count (P = 0.007) and was independently associated with shorter overall survival (Hazard Ratio: 3.6; 95% Confidence Interval: 1.17-11.28; P = 0.026). Taken together, our data highlights the importance of NTAL in APL cell survival and response to treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Leukemia, Promyelocytic, Acute/pathology , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Adult , Aged , Animals , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease-Free Survival , Female , Gene Knockdown Techniques , Humans , Leukemia, Promyelocytic, Acute/blood , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/mortality , Leukocyte Count , Male , Membrane Microdomains/metabolism , Mice , Middle Aged , Retrospective Studies , Survival Analysis , Tretinoin/pharmacology , Tretinoin/therapeutic use , Xenograft Model Antitumor Assays , Young Adult
6.
J Proteomics ; 219: 103734, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32201364

ABSTRACT

Epithelial to Mesenchymal Transition (EMT) is a normal cellular process that is also triggered during cancer progression and metastasis. EMT induces cellular and microenviromental changes, resulting in loss of epithelial features and acquisition of mesenchymal phenotypes. The growth factor TGFß and the transcription factor SNAIL1 (SNAIL) have been described as inducers of EMT. Here, we carried out an EMT model with non-tumorigenic cell line MCF-10A induced with the TGFß2 specific isoform of TGF protein family. The model was validated by molecular, morphological and functional experiments and showed correlation with the up-regulation of SNAIL. In order to identify additional regulators of EMT in this non-tumorigenic model, we explored quantitative proteomics, which revealed the Ubiquitin carboxyl-terminal hydrolase 47 (USP47) as one of the top up-regulated proteins. USP47 has a known role in cell growth and genome integrity, but not previously correlated to EMT. After validating USP47 alterations using MRM and antibody-based assays, we demonstrated that the chemical inhibition of USP47 with the inhibitor P5091 reduced expression of EMT markers and reverted morphological changes in MCF-10A cells undergoing EMT. These results support the involvement of USP47 in our EMT model as well as potential applications of deubiquitinases as therapeutic targets for cancer progression management. BIOLOGICAL SIGNIFICANCE: Metastasis is responsible for most cancer-associated mortality. Additionally, metastasis requires special attention, as the cellular transformations make treatment at this stage very difficult or occasionally impossible. Early steps in cancer metastasis involve the ability to detach from the solid tumor mass and invade the surrounding stromal tissues through cohesive migration, or a mesenchymal or amoeboid invasion. One of the first steps for metastatic cascade is denominated epithelial to mesenchymal transition (EMT), which can be triggered by different factors. Here, our efforts were directed to better understand this process and identify new pathways that contributes for acquisition of EMT, mainly focused on post translational modifications related to ubiquitin proteasome system. Our model of EMT induction by TGFß2 mimics early stage of metastatic cancer in epithelial breast cells and a proteomic study carried out for such model demonstrates that the deubiquitinase enzyme USP47 acts in SNAIL stabilization, one of the most important transcription factors for EMT phenotype acquisition and consequent metastasis. In addition, the inhibiton of USP47 with P5091, reverted the EMT phenotype. Together the knowledge of such processes of cancer progression and regulation can help in designing new strategies for combined therapies for control of cancer in early stages.


Subject(s)
Epithelial-Mesenchymal Transition , Proteomics , Cell Line, Tumor , Cell Movement , Humans , Neoplasm Invasiveness , Transcription Factors , Transforming Growth Factor beta2 , Ubiquitin Thiolesterase , Ubiquitin-Specific Proteases
7.
Mol Omics ; 15(5): 316-330, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31429845

ABSTRACT

The main cause of death in cancer is the spread, or metastasis, of cancer cells to distant organs with consequent tumor formation. Additionally, metastasis is a process that demands special attention, as the cellular transformations make cancer at this stage very difficult or occasionally even impossible to be cured. The main process that converts epithelial tumor cells to mesenchymal-like metastatic cells is the Epithelial to Mesenchymal Transition (EMT). This process allows stationary and polarized epithelial cells, which are connected laterally to several types of junctions as well as the basement membrane, to undergo multiple biochemical changes that enable disruption of cell-cell adherence and apical-basal polarity. Moreover, the cells undergo important reprogramming to remodel the cytoskeleton and acquire mesenchymal characteristics such as enhanced migratory capacity, invasiveness, elevated resistance to apoptosis and a large increase in the production of ECM components. As expected, the alterations of the protein complement are extensive and complex, and thus exploring this by proteomic approaches is of particular interest. Here we review the overall findings of proteome modifications during EMT, mainly focusing on molecular signatures observed in multiple proteomic studies as well as coordinated pathways, cellular processes and their clinical relevance for altered proteins. As a result, an interesting set of proteins is highlighted as potential targets to be further investigated in the context of EMT, metastasis and cancer progression.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Proteome , Proteomics , Cell Transformation, Neoplastic/metabolism , Epithelial Cells , Gene Expression Regulation, Neoplastic , Humans
8.
Article in English | MEDLINE | ID: mdl-31297369

ABSTRACT

The therapeutic potential of mesenchymal stem/stromal cells (MSC) is widely recognized for the treatment of several diseases, including acute graft-vs.-host disease (GVHD), hematological malignancies, cardiovascular, bone, and cartilage diseases. More recently, this therapeutic efficacy has been attributed to the bioactive molecules that these cells secrete (secretome), now being referred as medicinal signaling cells. This fact raises the opportunity of therapeutically using MSC-derived soluble factors rather than cells themselves, enabling their translation into the clinic. Indeed, many clinical trials are now studying the effects of MSC-secretome in the context of cell-free therapy. MSC secretome profile varies between donors, source, and culture conditions, making their therapeutic use very challenging. Therefore, identifying these soluble proteins and evaluating their production in a reproducible and scalable manner is even more relevant. In this work, we analyzed the global profile of proteins secreted by umbilical cord matrix (UCM) derived-MSC in static conditions by using mass spectrometry, enabling the identification of thousands of proteins. Afterwards, relevant proteins were chosen and monitored in the supernatant of a fully-controllable, closed and scalable system (bioreactor) by using multiple reaction monitoring (MRM) mass spectrometric technique in a time-dependent manner. The results showed that the majority of interesting proteins were enriched through time in culture, with the last day of culture being the ideal time for supernatant collection. The use of this regenerative "soup," which is frequently discarded, could represent a step toward a safe, robust and reproducible cell-free product to be used in the medical therapeutic field. The future use of chemically defined culture-media will certainly facilitate secretome production according to Good Manufacturing Practice (GMP) standards.

9.
Leuk Lymphoma ; 60(11): 2658-2668, 2019 11.
Article in English | MEDLINE | ID: mdl-31060403

ABSTRACT

Lipid rafts are ordered membrane domains, which provide an environment for the proteins participating in signal transduction. Perifosine is an alkylphospholipid (APL) that inhibits the AKT pathway, cytotoxic to neoplastic cells. We have shown that the lipid raft adaptor protein NTAL is a target of APLs in leukemic cells. Using human mantle cell lymphoma (MCL) Granta-519 cell line we showed here that perifosine decreased NTAL in lipid raft fractions reducing AKT phosphorylation before apoptosis. We also showed that the NTAL-knockdown by shRNA induced a state of reduced AKT activation. Experimental NTAL-knockdown in NSG mouse MCL xenografts reduced AKT activity, increased the basal apoptotic rate by 3-fold (n = 8) and decreased tumor weight by 2.7-fold (n = 5), indicating that NTAL participates in tumor growth. NTAL protein was detected by western blotting in circulating cells of 7 of 8 MCL patients in the leukemic phase, suggesting involvement in the progression of the disease.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Lymphoma, Mantle-Cell/pathology , Proto-Oncogene Proteins c-akt/metabolism , Adaptor Proteins, Signal Transducing/genetics , Aged , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Humans , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Prognosis , Proto-Oncogene Proteins c-akt/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Cytotherapy ; 18(7): 846-59, 2016 07.
Article in English | MEDLINE | ID: mdl-27260206

ABSTRACT

Multipotent mesenchymal stromal cells (MSC) are imbued with an immunosuppressive phenotype that extends to several immune system cells. In this study, we evaluated how distinct Toll-like receptor (TLR) agonists impact immunosuppressive properties of bone marrow (BM)-MSC and explored the potential mechanisms involved. We show that TLR4 stimulation by lipopolysaccharide (LPS) restricted the ability of MSC to suppress the proliferation of T lymphocytes, increasing the gene expression of interleukin (IL)-1ß and IL-6. In contrast, stimulation of TLR9 by DSP30 induced proliferation and the suppressive potential of BM-MSC, coinciding with reducing tumor necrosis factor (TNF)-α expression, increased expression of transforming growth factor (TGF)-ß1, increased percentages of BM-MSC double positive for the ectonucleotidases CD39+CD73+ and adenosine levels. Importantly, following simultaneous stimulation with LPS and DSP30, BM-MSC's ability to suppress T lymphocyte proliferation was comparable with that of non-stimulated BM-MSC levels. Moreover, stimulation of BM-MSC with LPS reduced significantly the gene expression levels, on co-cultured T lymphocyte, of IL-10 and interferon (IFN)γ, a cytokine with potential to enhance the immunosuppression mediated by MSC and ameliorate the clinical outcome of patients with graft-versus-host disease (GVHD). Altogether, our findings reiterate the harmful effects of LPS on MSC immunosuppression, besides indicating that DSP30 could provide a protective effect against LPS circulating in the blood of GVHD patients who receive BM-MSC infusions, ensuring a more predictable immunosuppressive effect. The novel effects and potential mechanisms following the stimulation of BM-MSC by DSP30 might impact their clinical use, by allowing the derivation of optimal "licensing" protocols for obtaining therapeutically efficient MSC.


Subject(s)
Adenosine/pharmacology , Immunosuppressive Agents/pharmacology , Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/cytology , Antigens, CD/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Coculture Techniques , Gene Expression Regulation/drug effects , Humans , Immunosuppression Therapy , Ligands , Lymphocyte Activation/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Oligonucleotides/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptors/metabolism
11.
Arch Insect Biochem Physiol ; 93(1): 3-24, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27160491

ABSTRACT

The integument of insects and other arthropods is composed of an inner basal lamina coated by the epidermis, which secretes the bulk of the outer integument layer, the cuticle. The genome sequencing of several insect species has allowed predicting classes of proteins integrating the cuticle. However, only a small proportion of them, as well as other proteins in the integumentary system, have been validated. Using two-dimensional gel electrophoresis coupled with mass spectrometry, we identified 45 different proteins in a total of 112 selected gel spots derived from thoracic integument samples of developing honeybee workers, including 14 cuticular proteins (AmelCPR 3, AmelCPR 12, AmelCPR 16, AmelCPR 27, apidermin 2, apidermin 3, endocuticle structural glycoprotein SgAbd-8-like, LOC100577363, LOC408365, LOC413679, LOC725454, LOC100576916, LOC725838, and peritrophin 3-C analogous). Gene ontology functional analysis revealed that the higher proportions of the identified proteins have molecular functions related to catalytic and structural molecule activities, are involved in metabolic biological processes, and pertain to the protein class of structural or cytoskeletal proteins and hydrolases. It is noteworthy that 26.7% of the identified proteins, including five cuticular proteins, were revealed as protein species resulting from allelic isoforms or derived from posttranslational modifications. Also, 66.7% of the identified cuticular proteins were expressed in more than one developmental phase, thus indicating that they are part of the larval, pupal, and adult cuticle. Our data provide experimental support for predicted honeybee gene products and new information on proteins expressed in the developing integument.


Subject(s)
Bees/genetics , Gene Expression , Insect Proteins/genetics , Animals , Bees/growth & development , Bees/metabolism , Electrophoresis, Gel, Two-Dimensional , Insect Proteins/metabolism , Integumentary System/physiology , Larva/genetics , Larva/growth & development , Larva/metabolism , Pupa/genetics , Pupa/growth & development , Pupa/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
Mol Cell Proteomics ; 15(3): 906-17, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26764010

ABSTRACT

Epithelial to mesenchymal transition (EMT)(1) occurs naturally during embryogenesis, tissue repair, cancer progression, and metastasis. EMT induces cellular and microenvironmental changes resulting in loss of epithelial and acquisition of mesenchymal phenotypes, which promotes cellular invasive and migratory capabilities. EMT can be triggered by extracellular factors, including TGF-ß, HGF, and EGF. Overexpression of transcription factors, such as SNAIL, SLUG, ZEB1/2, and TWIST1, also induces EMT and is correlated to cancer aggressiveness. Here, the breast adenocarcinoma cell line MCF7 was transduced with SNAIL to identify specific mechanisms controlled by this transcription factor during EMT. Overexpression of SNAIL led to EMT, which was thoroughly validated by molecular, morphological, and functional experiments. Subcellular proteome enrichment followed by GEL-LC-MS/MS was performed to provide extensive protein fractionation and in-depth proteomic analysis. Quantitative analysis relied on a SILAC strategy, using the invasive breast cancer cell line MDA-MB-231 as a reference for quantitation. Subsets of proteins enriched in each subcellular compartment led to a complementary list of 4289 proteins identified with high confidence. A subset of differentially expressed proteins was validated by Western blot, including regulation in specific cellular compartments, potentially caused by protein translocation. Protein network analysis highlighted complexes involved in cell cycle control and epigenetic regulation. Flow cytometry analysis indicated that SNAIL overexpression led to cell cycle arrest in G0/G1 phases. Furthermore, down-regulation of HDAC1 was observed, supporting the involvement of epigenetic processes in SNAIL-induced EMT. When HDAC1 activity was inhibited, MCF7 not only apparently initiated EMT but also up-regulated SNAIL, indicating the cross-talk between these two proteins. Both HDAC1 inhibition and SNAIL overexpression activated the AKT pathway. These molecular mechanisms appear to be essential to EMT and therefore for cancer metastasis. Specific control of such epigenetic processes might then represent effective approaches for clinical management of metastatic cancer.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Histone Deacetylase 1/metabolism , Proteomics/methods , Snail Family Transcription Factors/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Epigenesis, Genetic , Female , Gene Regulatory Networks , Humans , MCF-7 Cells , Neoplasm Invasiveness , Proteome/isolation & purification , Proteome/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...