Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2817: 33-43, 2024.
Article in English | MEDLINE | ID: mdl-38907145

ABSTRACT

Mass spectrometry-based proteomics has traditionally been limited by the amount of input material for analysis. Single-cell proteomics has emerged as a challenging discipline due to the ultra-high sensitivity required. Isobaric labeling-based multiplex strategies with a carrier proteome offer an approach to overcome the sensitivity limitations. Following this as the basic strategy, we show here the general workflow for preparing cells for single-cell mass spectrometry-based proteomics. This protocol can also be applied to manually isolated cells when large cells, such as cardiomyocytes, are difficult to isolate properly with conventional fluorescence-activated cell sorting (FACS) sorter methods.


Subject(s)
Proteomics , Single-Cell Analysis , Proteomics/methods , Single-Cell Analysis/methods , Humans , Mass Spectrometry/methods , Flow Cytometry/methods , Proteome/analysis , Animals , Isotope Labeling/methods , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/cytology , Staining and Labeling/methods
2.
Int J Mol Sci ; 23(22)2022 Nov 14.
Article in English | MEDLINE | ID: mdl-36430510

ABSTRACT

Protein kinase C (PKC) comprises a family of highly related serine/threonine protein kinases involved in multiple signaling pathways, which control cell proliferation, survival, and differentiation. The role of PKCα in cancer has been studied for many years. However, it has been impossible to establish whether PKCα acts as an oncogene or a tumor suppressor. Here, we analyzed the importance of PKCα in cellular processes such as proliferation, migration, or apoptosis by inhibiting its gene expression in a luminal A breast cancer cell line (MCF-7). Differential expression analysis and phospho-kinase arrays of PKCα-KD vs. PKCα-WT MCF-7 cells identified an essential set of proteins and oncogenic kinases of the JAK/STAT and PI3K/AKT pathways that were down-regulated, whereas IGF1R, ERK1/2, and p53 were up-regulated. In addition, unexpected genes related to the interferon pathway appeared down-regulated, while PLC, ERBB4, or PDGFA displayed up-regulated. The integration of this information clearly showed us the usefulness of inhibiting a multifunctional kinase-like PKCα in the first step to control the tumor phenotype. Then allowing us to design a possible selection of specific inhibitors for the unexpected up-regulated pathways to further provide a second step of treatment to inhibit the proliferation and migration of MCF-7 cells. The results of this study suggest that PKCα plays an oncogenic role in this type of breast cancer model. In addition, it reveals the signaling mode of PKCα at both gene expression and kinase activation. In this way, a wide range of proteins can implement a new strategy to fine-tune the control of crucial functions in these cells and pave the way for designing targeted cancer therapies.


Subject(s)
Neoplasms , Protein Kinase C-alpha , Humans , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Protein Kinase C/metabolism , Cell Proliferation
3.
Proteomics Clin Appl ; 14(1): e1900052, 2020 01.
Article in English | MEDLINE | ID: mdl-31502404

ABSTRACT

PURPOSE: Successful prevention of colorectal cancer (CRC) would benefit from a rapid serum screening for early detection. Here, a novel strategy for CRC biomarker discovery and validation exclusively based on MS procedures is reported. EXPERIMENTAL DESIGN: Identification of CRC serum biomarkers is initially made using label-free quantification on pooled serum samples from different CRC stages followed by two consecutive steps of targeted parallel reaction monitoring assays in different serum cohorts. Relevance of different protein depletion and peptide fractionation extent is investigated. Absolute quantification of a selected peptide is performed as a proof-of-concept. RESULTS: A total of 945 proteins showed differential abundance in the discovery phase. Based on their statistical significance and relative expression in disease stages, 123 potential biomarkers are selected for a training step. In the final validation step, five peptides belonging to four proteins are consistently quantified in individual CRC serum samples and controls. Different statistical analyses indicate that peptides GWVTDGFSSLK (APOC3) and LCNNPTPQFGGK (THBS1) are candidate biomarkers. Absolute quantification of LCNNPTPQFGGK shows statistical significance for the diagnosis of early respect to late CRC stages. CONCLUSIONS AND CLINICAL RELEVANCE: Two peptides from APOC3 and THBS1 are validated by PRM as potential biomarkers for non-invasive diagnosis of colorectal cancer.


Subject(s)
Apolipoprotein C-III/blood , Biomarkers, Tumor/blood , Colorectal Neoplasms/blood , Peptides/blood , Thrombospondins/blood , Colorectal Neoplasms/pathology , Early Detection of Cancer , Female , Humans , Male , Neoplasm Proteins/blood , Proteome/genetics
4.
Inorg Chem ; 58(19): 12954-12963, 2019 Oct 07.
Article in English | MEDLINE | ID: mdl-31550148

ABSTRACT

The luminescent chalcone gold(I) conjugates [Au(PPh3)(AN3E)]PF6(1) and [Au(SIMes)(AN3E)]PF6 (2) (AN3E = (E)-3-(9-anthracenyl)-1-(4-pyridyl)propenone; SIMes = N,N'-dimesitylimidazolidin-2-ylidene; Mes = 2,4,6-trimethylphenyl)) were prepared and characterized; complex 1 was also characterized by X-ray crystallography. In MTT assays against a panel of three human colon, a melanoma and a breast cancer cell lines both complexes were antiproliferative with low micromolar IC50 values. It is noteworthy that HCT116p53-/- colon carcinoma cells lacking functional p53 (a vital tumor suppressor) were more susceptible to them than the wildtype parent cell line. In flow cytometry analyses, the gold conjugates induced a significant arrest in G2/M phase primarily. Complexes 1 and 2 quickly increased the production of reactive oxygen species (ROS) and induced mitochondrial membrane potential depolarization, higher ROS values being obtained after coadministration with enzymatic inhibitors. The free chalcone AN3E and its gold(I) complex conjugates located in the cell mitochondria according to confocal microscopy. In addition, complexes 1 and 2 showed in vivo antivascular effects on the chorioallantoic membrane (CAM) of fertilized specific-pathogen-free (SPF) chicken eggs.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Anthracenes/pharmacology , Antineoplastic Agents/pharmacology , Chalcone/pharmacology , Colonic Neoplasms/drug therapy , Organogold Compounds/pharmacology , Angiogenesis Inhibitors/chemistry , Animals , Anthracenes/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Proliferation/drug effects , Chalcone/analogs & derivatives , Chickens , Crystallography, X-Ray , HCT116 Cells , Humans , Models, Molecular , Organogold Compounds/chemistry
5.
J Proteome Res ; 17(1): 252-264, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29131639

ABSTRACT

We investigated new transcription and splicing factors associated with the metastatic phenotype in colorectal cancer. A concatenated tandem array of consensus transcription factor (TF)-response elements was used to pull down nuclear extracts in two different pairs of colorectal cancer cells, KM12SM/KM12C and SW620/480, genetically related but differing in metastatic ability. Proteins were analyzed by label-free LC-MS and quantified with MaxLFQ. We found 240 proteins showing a significant dysregulation in highly metastatic KM12SM cells relative to nonmetastatic KM12C cells and 257 proteins in metastatic SW620 versus SW480. In both cell lines there were similar alterations in genuine TFs and components of the splicing machinery like UPF1, TCF7L2/TCF-4, YBX1, or SRSF3. However, a significant number of alterations were cell-line specific. Functional silencing of MAFG, TFE3, TCF7L2/TCF-4, and SRSF3 in KM12 cells caused alterations in adhesion, survival, proliferation, migration, and liver homing, supporting their role in metastasis. Finally, we investigated the prognostic value of the altered TFs and splicing factors in cancer patients. SRSF3 and SFPQ showed significant prognostic value. We observed that SRSF3 displayed a gradual loss of expression associated with cancer progression. Loss of SRSF3 expression was significantly associated with poor survival and shorter disease-free survival, particularly in early stages, in colorectal cancer.


Subject(s)
Colorectal Neoplasms/chemistry , Neoplasm Metastasis , Proteomics/methods , RNA Splicing Factors/analysis , Transcription Factors/analysis , Cell Line, Tumor , Colorectal Neoplasms/pathology , Humans , Prognosis , Serine-Arginine Splicing Factors/analysis
6.
J Proteome Res ; 16(12): 4374-4390, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28960077

ABSTRACT

The Human Proteome Project (HPP) aims deciphering the complete map of the human proteome. In the past few years, significant efforts of the HPP teams have been dedicated to the experimental detection of the missing proteins, which lack reliable mass spectrometry evidence of their existence. In this endeavor, an in depth analysis of shotgun experiments might represent a valuable resource to select a biological matrix in design validation experiments. In this work, we used all the proteomic experiments from the NCI60 cell lines and applied an integrative approach based on the results obtained from Comet, Mascot, OMSSA, and X!Tandem. This workflow benefits from the complementarity of these search engines to increase the proteome coverage. Five missing proteins C-HPP guidelines compliant were identified, although further validation is needed. Moreover, 165 missing proteins were detected with only one unique peptide, and their functional analysis supported their participation in cellular pathways as was also proposed in other studies. Finally, we performed a combined analysis of the gene expression levels and the proteomic identifications from the common cell lines between the NCI60 and the CCLE project to suggest alternatives for further validation of missing protein observations.


Subject(s)
Proteome/analysis , Proteomics/methods , Search Engine , Cell Line, Tumor , Humans , Knowledge Bases , Proteins/analysis , Software
7.
J Chem Theory Comput ; 13(8): 3898-3910, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28641006

ABSTRACT

In this work, we use DFT-based methods to simulate the chemical structures, optical properties, and interaction with DNA of a recently synthesized chelated C^N 9-aminoacridine arene Ru(II) anticancer agent and two new closely related Rh(III) and Ir(III) complexes using DFT-based methods. Four chemical models and a number of theoretical approaches, which representatively include the PBE0, B97D, ωB97X, ωB97X-D, M06, and M06-L density functionals and the LANL2DZ, def2-SVP, and def2-TZVP basis sets, are tested. The best overall accuracy/cost performance for the optimization process is reached at the ωB97X-D/def2-SVP and M06/def2-SVP levels of theory. Inclusion of explicit solvent molecules (CHCl3) further refines the geometry, while taking into account the crystal network gives no significant improvements of the computed bond distances and angles. The analysis of the excited states reveals that the M06 level matches better the experimental absorption spectra, compared to ωB97X-D. The use of the M06/def2-SVP approach is therefore a well-balanced method to study theoretically the bioactivity of this type of antitumoral complexes, so we couple this TD-DFT approach to molecular dynamics simulations in order to assess their reactivity with DNA. The reported results demonstrate that these drugs could be used to inject electrons into DNA, which might broaden their applications in photoactivated chemotherapy and as new materials for DNA-based electrochemical nanodevices.


Subject(s)
Aminacrine/analogs & derivatives , Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , DNA/chemistry , Intercalating Agents/chemistry , Electrons , Iridium/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Quantum Theory , Rhodium/chemistry , Ruthenium/chemistry
8.
ACS Chem Biol ; 12(6): 1524-1537, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28388047

ABSTRACT

Two new 1-acridin-9-yl-3-methylthiourea Au(I) DNA intercalators [Au(ACRTU)2]Cl (2) and [Au(ACRTU) (PPh3)]PF6 (3) have been prepared. Both complexes were highly active in the human ovarian carcinoma cisplatin-sensitive A2780 cell line, exhibiting IC50 values in the submicromolar range. Compounds 2 and 3 are also cytotoxic toward different phenotypes of breast cancer cell lines MDA-MB-231 (triple negative), SK-BR-3 (HER2+, ERα-, and ERß-), and MCF-7 (ER+). Both complexes induce apoptosis through activation of caspase-3 in vitro. While inhibition of some proteins (thiol-containing enzymes) seems to be the main mechanism of action for cytotoxic gold complexes, 2 and 3 present a DNA-dependent mechanism of action. They locate in the cell nucleus according to confocal microscopy and transmission electronic microscopy. The binding to DNA resulted to be via intercalation as shown by spectroscopic methods and viscometry, exhibiting a dose-dependent response on topoisomerase I mediated DNA unwinding. In addition, 2 and 3 exhibit potent antiangiogenic effects and are also able to inhibit vasculogenic mimicry of highly invasive MDA-MB-231 cells.


Subject(s)
Antineoplastic Agents/chemistry , Intercalating Agents/chemistry , Acridines/chemistry , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Nucleus/metabolism , Gold/chemistry , Humans , Intercalating Agents/pharmacology , Molecular Mimicry , Neoplasm Invasiveness/prevention & control , Neovascularization, Pathologic/drug therapy , Thiourea/chemistry
9.
Sci Rep ; 5: 11176, 2015 Jun 08.
Article in English | MEDLINE | ID: mdl-26052917

ABSTRACT

Phenomenological screening of small molecule libraries for anticancer activity yields potentially interesting candidate molecules, with a bottleneck in the determination of drug targets and the mechanism of anticancer action. We have found that, for the protein target of a small-molecule drug, the abundance change in late apoptosis is exceptional compared to the expectations based on the abundances of co-regulated proteins. Based on this finding, a novel method to drug target deconvolution is proposed. In a proof of principle experiment, the method yielded known targets of several common anticancer agents among a few (often, just one) likely candidates identified in an unbiased way from cellular proteome comprising more than 4,000 proteins. A validation experiment with a different set of cells and drugs confirmed the findings. As an additional benefit, mapping most specifically regulated proteins on known protein networks highlighted the mechanism of drug action. The new method, if proven to be general, can significantly shorten drug target identification, and thus facilitate the emergence of novel anticancer treatments.


Subject(s)
Antineoplastic Agents/pharmacology , Computational Biology/methods , Drug Discovery/methods , Proteome/drug effects , Small Molecule Libraries/pharmacology , Apoptosis/drug effects , Camptothecin/pharmacology , Cell Line, Tumor , Doxorubicin/pharmacology , Fluorouracil/pharmacology , HCT116 Cells , Humans , Methotrexate/pharmacology , Paclitaxel/pharmacology , Proteomics , Quinazolines/pharmacology , Thiophenes/pharmacology
10.
J Cell Sci ; 128(6): 1097-107, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25632158

ABSTRACT

The exosome acts on different RNA substrates and plays important roles in RNA metabolism. The fact that short non-coding RNAs are involved in the DNA damage response led us to investigate whether the exosome factor RRP6 of Drosophila melanogaster and its human ortholog EXOSC10 play a role in DNA repair. Here, we show that RRP6 and EXOSC10 are recruited to DNA double-strand breaks (DSBs) in S2 cells and HeLa cells, respectively. Depletion of RRP6/EXOSC10 does not interfere with the phosphorylation of the histone variant H2Av (Drosophila) or H2AX (humans), but impairs the recruitment of the homologous recombination factor RAD51 to the damaged sites, without affecting RAD51 levels. The recruitment of RAD51 to DSBs in S2 cells is also inhibited by overexpression of RRP6-Y361A-V5, a catalytically inactive RRP6 mutant. Furthermore, cells depleted of RRP6 or EXOSC10 are more sensitive to radiation, which is consistent with RRP6/EXOSC10 playing a role in DNA repair. RRP6/EXOSC10 can be co-immunoprecipitated with RAD51, which links RRP6/EXOSC10 to the homologous recombination pathway. Taken together, our results suggest that the ribonucleolytic activity of RRP6/EXOSC10 is required for the recruitment of RAD51 to DSBs.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Homologous Recombination/genetics , Animals , Blotting, Western , Cell Proliferation , Chromatin Immunoprecipitation , Drosophila Proteins/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Exoribonucleases/antagonists & inhibitors , Exoribonucleases/genetics , Exosome Multienzyme Ribonuclease Complex/antagonists & inhibitors , Exosome Multienzyme Ribonuclease Complex/genetics , HeLa Cells , Histones/metabolism , Humans , Phosphorylation , RNA, Small Interfering/genetics , Rad51 Recombinase/metabolism
11.
J Proteome Res ; 12(4): 1969-79, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23477467

ABSTRACT

The pyrimidine analogue 5-fluorouracil (5FU) is used as a treatment for solid tumors, but its mechanism of action is not fully understood. We have used mass spectrometry to study the mechanism of action of 5FU, and we have measured the effects of this drug on the composition and on the turnover of the proteome of RKO cancer cells. We have identified novel potential targets of 5FU that are affected after very short exposure times. We have also shown that 5FU has a massive effect on the proteins involved in RNA metabolism. After only 1 h of treatment, 5FU causes a post-transcriptional reduction in the abundance of components of the translation machinery (mostly ribosomal proteins), and this reduction is accompanied by a down-regulation of the translational capacity of the cells. Neither rapamycin nor raltitrexed, two drugs that also block cell proliferation, reduce the abundances of ribosomal proteins as 5FU does, which suggests that the down-regulation of ribosomal proteins is coupled to the mechanism of action of 5FU. Some of our observations conflict with previous reports based on RNA quantification. This shows how important it is to complement RNA profiling studies with analyses of drug toxicity at the protein level.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Fluorouracil/pharmacology , Proteome/metabolism , Cell Line, Tumor , Colonic Neoplasms/genetics , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Protein Biosynthesis/drug effects , Ribosomal Proteins/metabolism , Tandem Mass Spectrometry
12.
Inorg Chem ; 52(2): 974-82, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23301676

ABSTRACT

A series of new organoiridium(III) complexes [Ir(N-C)(2)(N-S)]Cl (HN-C = 2-phenylpyridine (Hppy), N-S = methyl thiosemicarbazide (1), phenyl thiosemicarbazide (2) and naphtyl thiosemicarbazide (3)) have been synthesized and characterized. The crystal structure of (1) has been established by X-ray diffraction, showing the thiosemicarbazide ligand bound to the iridium atom as N,S-chelate. The cytotoxicity studies show that they are more active than cisplatin (about 5-fold) in T47D (breast cancer) at 48 h incubation time. On the other hand, very low resistance factors (RF) of 1-3 in A2780cisR (cisplatin-resistant ovarian carcinoma) at 48 h were observed (RF ≈ 1). Ir accumulation in T47D cell line after 48 h continuous exposure for complexes 1-3 are higher than that corresponding to cisplatin (about 10 times). The complexes 1-3 bind strongly to HSA with binding constants of about 10(4) M(-1) at 296 K, binding occurring at the warfarin site I for 2. Complexes 2 and 3 are also capable of binding in the minor groove of DNA as shown by Hoechst 33258 displacement experiments. Furthermore, complex 2 is also a good cathepsin B inhibitor (an enzyme implicated in a number of cancer related events), being the enzyme reactivated by cysteine.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cathepsin B/antagonists & inhibitors , DNA/chemistry , Iridium/chemistry , Semicarbazides/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding, Competitive , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , DNA/metabolism , Electrophoresis, Gel, Two-Dimensional , Enzyme Activation/drug effects , Female , Humans , Inhibitory Concentration 50 , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Ovarian Neoplasms/drug therapy , Semicarbazides/chemical synthesis , Semicarbazides/pharmacology , Serum Albumin/chemistry , Serum Albumin/metabolism
13.
Dalton Trans ; 41(1): 300-6, 2012 Jan 07.
Article in English | MEDLINE | ID: mdl-22089173

ABSTRACT

The interaction of a novel Pt complex, [Pt(dmba)(N9-9AA)(PPh(3))](+)1 (dmba = N,N-dimethylbenzylamine-κN,κC; 9AA = 9-aminoacridine), which exhibits anti-tumor activity, with certain key proteins has been monitored by ESI-MS. Also, the interaction of 1 with a designed double-stranded oligonucleotide containing the GG motif has been followed by mass spectrometry as well as by fluorimetry. The results obtained show the low interaction of 1 with the considered proteins and the absence of covalent interaction with the oligonucleotides, but the fluorimetric data confirm the π-π interaction of 1 with the double-stranded DNA, which is probably the reason of the previously reported activity of 1 in several tumor cell lines.


Subject(s)
Aminacrine/chemistry , Antineoplastic Agents/chemistry , Coordination Complexes/chemistry , Oligonucleotides/chemistry , Platinum/chemistry , Proteins/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/toxicity , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Coordination Complexes/toxicity , Humans , Spectrometry, Mass, Electrospray Ionization
14.
Inorg Chem ; 50(6): 2151-8, 2011 Mar 21.
Article in English | MEDLINE | ID: mdl-21314142

ABSTRACT

The synthesis of the novel anthraquinone platinum derivate [Pt(ppy)Cl(1C3)] (2) [Hppy = N,C-chelating 2-phenylpyridine; 1C3 = 1-[(3-aminopropyl)amino]-anthracene-9,10-dione] and its values of IC(50) against a panel of human tumor cell lines representative of ovarian (A2780 and A2780cisR) and breast cancers (T47D) are reported. At 24 h incubation time, complex 2 was more active than cisplatin (about 9-fold) and the free ligand 1C3 (about 2-fold) in T47-D. The observation that the cisplatin IC(50) falls by about 10-fold from 24 to 72 h, whereas that for 2 changes little, suggests substantial differences in the mode of action. Complex 2 also showed high cytotoxicity against A2780 (about 3-fold greater than cisplatin at 24 h). On the other hand, very low resistance factors (RF) of 2 in A2780cisR at 24-72 h (RF = 1.3) were observed. The interaction of 2 with DNA was followed by electrophoretic mobility and UV-visible spectroscopy, and its reaction with the model nucleobase 9-EtG was studied by (1)H NMR and ESI-MS. Theoretical calculations at the B3LYP/def2-TZVPP//BP86/def2-TZVP level of theory on complex 2 show a labile Pt-Cl bond that allows easy replacement of Cl by N-nucleophiles such as 9-EtG, which forms a stronger Pt-N bond.


Subject(s)
Anthraquinones/chemistry , Antineoplastic Agents/pharmacology , Organoplatinum Compounds/pharmacology , Pyridines/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Models, Molecular , Molecular Structure , Organoplatinum Compounds/chemical synthesis , Organoplatinum Compounds/chemistry , Quantum Theory , Structure-Activity Relationship , Tumor Cells, Cultured
15.
J Proteome Res ; 10(2): 529-40, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-20973479

ABSTRACT

PKCα is a key mediator of the neuronal differentiation controlled by NGF and ATP. However, its downstream signaling pathways remain to be elucidated. To identify the signaling partners of PKCα, we analyzed proteins coimmunoprecipitated with this enzyme in PC12 cells differentiated with NGF and ATP and compared them with those obtained with NGF alone or growing media. Mass spectrometry analysis (LC-MS/MS) identified plectin, peripherin, filamin A, fascin, and ß-actin as potential interacting proteins. The colocalization of PKCα and its interacting proteins increased when PC12 cells were differentiated with NGF and ATP. Peripherin and plectin organization and the cortical remodeling of ß-actin were dramatically affected when PKCα was down-regulated, suggesting that all three proteins might be functional targets of ATP-dependent PKCα signaling. Taken together, these data demonstrate that PKCα is essential for controlling the neuronal development induced by NGF and ATP and interacts with the cytoskeletal components at two levels: assembly of the intermediate filament peripherin and organization of cortical actin.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Differentiation/physiology , Cytoskeletal Proteins/metabolism , Neurons/metabolism , Protein Kinase C-alpha/metabolism , Animals , Blotting, Western , Down-Regulation , Intermediate Filament Proteins/metabolism , Intermediate Filaments/metabolism , Intracellular Space/metabolism , Membrane Glycoproteins/metabolism , Microscopy, Fluorescence , Models, Biological , Nerve Growth Factor/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Neurons/cytology , Neurons/enzymology , PC12 Cells , Peripherins , Plectin/metabolism , Protein Interaction Mapping , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Rats , Reproducibility of Results , Signal Transduction/physiology , Tandem Mass Spectrometry
16.
Phys Chem Chem Phys ; 12(41): 13372-4, 2010 Nov 07.
Article in English | MEDLINE | ID: mdl-20856953

ABSTRACT

Detailed analysis of >18 400 high-mass accuracy tandem mass spectra resulting from higher energy collisional dissociation yields further evidence of the cyclic nature of a(2)-ions.


Subject(s)
Ions/chemistry , Diketopiperazines/chemistry , Oxazolone/chemistry , Tandem Mass Spectrometry
18.
Proc Natl Acad Sci U S A ; 106(16): 6603-7, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19346474

ABSTRACT

C2 domains are widely-spread protein signaling motifs that in classical PKCs act as Ca(2+)-binding modules. However, the molecular mechanisms of their targeting process at the plasma membrane remain poorly understood. Here, the crystal structure of PKCalpha-C2 domain in complex with Ca(2+), 1,2-dihexanoyl-sn-glycero-3-[phospho-L-serine] (PtdSer), and 1,2-diayl-sn-glycero-3-[phosphoinositol-4,5-bisphosphate] [PtdIns(4,5)P(2)] shows that PtdSer binds specifically to the calcium-binding region, whereas PtdIns(4,5)P(2) occupies the concave surface of strands beta3 and beta4. Strikingly, the structure reveals a PtdIns(4,5)P(2)-C2 domain-binding mode in which the aromatic residues Tyr-195 and Trp-245 establish direct interactions with the phosphate moieties of the inositol ring. Mutations that abrogate Tyr-195 and Trp-245 recognition of PtdIns(4,5)P(2) severely impaired the ability of PKCalpha to localize to the plasma membrane. Notably, these residues are highly conserved among C2 domains of topology I, and a general mechanism of C2 domain-membrane docking mediated by PtdIns(4,5)P(2) is presented.


Subject(s)
Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Kinase C-alpha/chemistry , Protein Kinase C-alpha/metabolism , Amino Acids/metabolism , Animals , Cations/metabolism , Cell Membrane/enzymology , Conserved Sequence , Genes, Dominant , Models, Molecular , Mutant Proteins/metabolism , PC12 Cells , Protein Structure, Secondary , Protein Structure, Tertiary , Protein Transport , Rats , Structure-Activity Relationship
19.
Inorg Chem ; 47(15): 6990-7001, 2008 Aug 04.
Article in English | MEDLINE | ID: mdl-18593114

ABSTRACT

The new complexes [Pd(dmba)( N10-9AA)(PPh 3)]ClO 4 ( 1), [Pt(dmba)( N9-9AA)(PPh 3)]ClO 4 ( 2), [Pd(dmba)( N10-9AA)Cl] ( 3), and [Pd(C 6F 5)( N10-9AA)(PPh 3)Cl] ( 4) (9-AA = 9-aminoacridine; dmba = N,C-chelating 2-(dimethylaminomethyl)phenyl) have been prepared. The crystal structures have been established by X-ray diffraction. In complex 2, an anagostic C-H...Pt interaction is observed. All complexes are luminescent in the solid state at room temperature, showing important differences between the palladium and platinum complexes. Complex 2 shows two structured emission bands at high and low energies in the solid state, and the lifetimes are in agreement with excited states of triplet parentage. Density functional theory and time-dependent density functional theory calculations for complex 2 have been done. Values of IC 50 were also calculated for the new complexes 1- 4 against the tumor cell line HL-60. All of the new complexes were more active than cisplatin (up to 30-fold in some cases). The DNA adduct formation of the new complexes synthesized was followed by circular dichroism and electrophoretic mobility. Atomic force microscopy images of the modifications caused by the complexes on plasmid DNA pB R322 were also obtained.


Subject(s)
Aminacrine/chemistry , Antineoplastic Agents/pharmacology , Luminescence , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Palladium/chemistry , Platinum/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chelating Agents , Circular Dichroism , Electrophoresis, Agar Gel , Humans , Microscopy, Atomic Force , Organometallic Compounds/chemistry , Quantum Theory , Time Factors
20.
J Mol Biol ; 377(4): 1038-52, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18304574

ABSTRACT

Rapamycin-triggered heterodimerization strategy is becoming an excellent tool for rapidly modifying phosphatidylinositol(4,5)-bisphosphate [PtdIns(4,5)P2] levels at the plasma membrane and for studying their influence in different processes. In this work, we studied the effect of modulation of the PtdIns(4,5)P2 concentration on protein kinase C (PKC) alpha membrane localization in intact living cells. We showed that an increase in the PtdIns(4,5)P2 concentration enlarges the permanence of PKCalpha in the plasma membrane when PC12 cells are stimulated with ATP, independently of the diacylglycerol generated. The depletion of this phosphoinositide decreases both the percentage of protein able to translocate to the plasma membrane and its permanence there. Our results demonstrate that the polybasic cluster located in the C2 domain of PKCalpha is responsible for this phosphoinositide-protein interaction. Furthermore, the C2 domain acts as a dominant interfering module in the neural differentiation process of PC12 cells, a fact that was also supported by the inhibitory effect obtained by knocking down PKCalpha with small interfering RNA duplexes. Taken together, these data demonstrate that PtdIns(4,5)P2 itself targets PKCalpha to the plasma membrane through the polybasic cluster located in the C2 domain, with this interaction being critical in the signaling network involved in neural differentiation.


Subject(s)
Cell Membrane/metabolism , Ligands , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Protein Kinase C-alpha/metabolism , Adenosine Triphosphate/pharmacology , Animals , Cell Differentiation/genetics , Cell Membrane/drug effects , Diglycerides/metabolism , Models, Biological , Nerve Growth Factor/pharmacology , Neurites/drug effects , PC12 Cells , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Binding , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/chemistry , Protein Kinase C-alpha/genetics , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Protein Transport/drug effects , RNA, Small Interfering/pharmacology , Rats , Sirolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...