Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Front Endocrinol (Lausanne) ; 12: 635405, 2021.
Article in English | MEDLINE | ID: mdl-34025576

ABSTRACT

Background: Cell therapy of diabetes aims at restoring the physiological control of blood glucose by transplantation of functional pancreatic islet cells. A potentially unlimited source of cells for such transplantations would be islet cells derived from an in vitro differentiation of human pluripotent stem cells (hESC/hiPSC). The islet-like clusters (ILC) produced by the known differentiation protocols contain various cell populations. Among these, the ß-cells that express both insulin and the transcription factor Nkx6.1 seem to be the most efficient to restore normoglycemia in diabetes animal models. Our aim was to find markers allowing selection of these efficient cells. Methods: Functional Cell-Capture Screening (FCCS) was used to identify markers that preferentially capture the cells expressing both insulin and Nkx6.1, from hESC-derived ILC cells. In order to test whether selection for such markers could improve cell therapy in diabetic mouse models, we used ILC produced from a clinical-grade line of hESC by a refined differentiation protocol adapted to up-scalable bioreactors. Re-aggregated MACS sorted cells were encapsulated in microspheres made of alginate modified to reduce foreign body reaction. Implantation was done intraperitoneally in STZ-treated C57BL/6 immuno-competent mice. Results: CD49A (integrin alpha1) was identified by FCCS as a marker for cells that express insulin (or C-peptide) as well as Nkx6.1 in ILC derived by hESC differentiation. The ILC fraction enriched in CD49A + cells rapidly reduced glycemia when implanted in diabetic mice, whereas mice receiving the CD49A depleted population remained highly diabetic. CD49A-enriched ILC cells also produced higher levels of human C-peptide in the blood of transplanted mice. However, the difference between CD49A-enriched and total ILC cells remained small. Another marker, CD26 (DPP4), was identified by FCCS as binding insulin-expressing cells which are Nkx6.1 negative. Depletion of CD26 + cells followed by enrichment for CD49A + cells increased insulin+/Nkx6.1+ cells fraction to ~70%. The CD26 - /CD49A + enriched ILC exhibited improved function over non-sorted ILC or CD49A + cells in diabetic mice and maintain prolonged blood C-peptide levels. Conclusions: Refining the composition of ILC differentiated from hPSC by negative selection to remove cells expressing CD26 and positive selection for CD49A expressing cells could enable more effective cell therapy of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Dipeptidyl Peptidase 4/biosynthesis , Integrin alpha1/biosynthesis , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Pluripotent Stem Cells/metabolism , Animals , C-Peptide/biosynthesis , Cell Differentiation , Cell Separation , Homeodomain Proteins/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred C57BL , Microspheres
2.
Methods Mol Biol ; 1487: 175-194, 2017.
Article in English | MEDLINE | ID: mdl-27924567

ABSTRACT

The ERK1 and ERK2 (ERK1/2) cascade is a central signaling pathway activated by a wide variety of extracellular agents that transmit the messages of G Protein Coupled Receptors (GPCRs) and Receptor Tyrosine Kinases (RTKs). Being such a central pathway, the activity of the cascade is well regulated, including by dynamic changes of the subcellular localization of components of the ERK1/2 cascade. In resting cells, ERK1/2 are localized in the cytosol due to their interactions with different anchoring proteins. After stimulation, ERK1/2 are phosphorylated by MEK1/2 on their regulatory TEY motif, which permits their detachment from the anchoring proteins. This detachment exposes ERK1/2 to additional phosphorylation on two serine residues (SPS motif) within the nuclear translocation signal (NTS) of the kinases. This additional phosphorylation allows ERK1/2 to interact with importin7, which consequently promotes their translocation to the nucleus. More studies are still required in order to better understand the mechanism and consequence of the nuclear translocation of ERK1/2. In this chapter, we describe some of the techniques used to study nuclear translocation of ERK1/2 in mammalian cells. We briefly mention methods such as digitonin permeabilization and cellular fractionation, as well as overexpression of reporter constructs. More thoroughly, we describe immunofluorescence, immunoprecipitation, and proximity ligation assay (PLA) approaches that are routinely used in our laboratory. Hopefully, the increase of knowledge based on these methods will open more opportunities for the identification of new therapeutic targets for diseases where the ERK1/2 cascade is dysregulated, such as cancer, neurodegenerative diseases, and diabetes.


Subject(s)
Cell Nucleus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorescent Antibody Technique , Carrier Proteins , Cell Cycle , Intracellular Space , Phosphorylation , Protein Binding , Protein Transport , Signal Transduction
3.
Nat Commun ; 6: 6685, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25819065

ABSTRACT

A hallmark of the ERK1/2 functioning is their nuclear translocation, which is mainly required for the induction of proliferation. Activated ERK1/2 molecules that remain in the cytoplasm initiate other activities, including immediate feedback loops. Prevention of the nuclear translocation should therefore inhibit proliferation, without affecting cytoplasm-induced cellular processes. Here we present an NTS-derived myristoylated phosphomimetic peptide, which blocks the interaction of importin7 and ERK1/2, and consequently the nuclear translocation of the latter. In culture, the peptide induces apoptosis of melanoma cells inhibits the viability of other cancer cells, but has no effect on non-transformed, immortalized cells. It even inhibits the viability of PLX4032- and U0126-resistant melanoma cells. In xenograft models, the peptide inhibits several cancers, and acts much better than PLX4032 in preventing melanoma recurrence. This study provides a proof of concept for using the nuclear translocation of ERK1/2 as a drug target for the combat of various ERK1/2-related cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Nucleus/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Peptides/pharmacology , Active Transport, Cell Nucleus/drug effects , Animals , Blotting, Western , CHO Cells , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/drug effects , Cricetulus , HCT116 Cells , HeLa Cells , Humans , Immunohistochemistry , Immunoprecipitation , In Situ Nick-End Labeling , Mice, Nude , Mice, SCID , Microscopy, Fluorescence , Molecular Targeted Therapy , Neoplasm Transplantation , Protein Transport/drug effects , Xenograft Model Antitumor Assays
4.
Proteomics ; 12(17): 2668-81, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22740317

ABSTRACT

Intracellular peptides generated by the proteasome and oligopeptidases have been suggested to function in signal transduction and to improve insulin resistance in mice fed a high-caloric diet. The aim of this study was to identify specific intracellular peptides in the adipose tissue of Wistar rats that could be associated with the physiological and therapeutic control of glucose uptake. Using semiquantitative mass spectrometry and LC/MS/MS analyses, we identified ten peptides in the epididymal adipose tissue of the Wistar rats; three of these peptides were present at increased levels in rats that were fed a high-caloric Western diet (WD) compared with rats fed a control diet (CD). The results of affinity chromatography suggested that in the cytoplasm of epididymal adipose tissue from either WD or CD rats, distinctive proteins bind to these peptides. However, despite the observed increase in the WD animals, the evaluated peptides increased insulin-stimulated glucose uptake in 3T3-L1 adipocytes treated with palmitate. Thus, intracellular peptides from the adipose tissue of Wistar rats can bind to specific proteins and facilitate insulin-induced glucose uptake in 3T3-L1 adipocytes.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/metabolism , Glucose/metabolism , Insulin Resistance , Peptides/analysis , Peptides/metabolism , 3T3 Cells , Adipocytes/cytology , Adipocytes/metabolism , Amino Acid Sequence , Animals , Chromatography, Affinity , Chromatography, Liquid , Energy Intake , Insulin/metabolism , Male , Mice , Molecular Sequence Data , Palmitic Acid/metabolism , Protein Binding , Proteins/metabolism , Rats , Rats, Wistar , Tandem Mass Spectrometry
5.
AAPS J ; 12(4): 608-16, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20665142

ABSTRACT

Cells produce and use peptides in distinctive ways. In the present report, using isotope labeling plus semi-quantitative mass spectrometry, we evaluated the intracellular peptide profile of TAP1/ß2m⁻(/)⁻ (transporter associated with antigen-processing 1/ß2 microglobulin) double-knockout mice and compared it with that of C57BL/6 wild-type animals. Overall, 92 distinctive peptides were identified, and most were shown to have a similar concentration in both mouse strains. However, some peptides showed a modest increase or decrease (~2-fold), whereas a glycine-rich peptide derived from the C-terminal of neurogranin (KGPGPGGPGGAGGARGGAGGGPSGD) showed a substantial increase (6-fold) in TAP1/ß2m⁻(/)⁻ mice. Thus, TAP1 and ß2microglobulin have a small influence on the peptide profile of neuronal tissue, suggesting that the presence of peptides derived from intracellular proteins in neuronal tissue is not associated with antigens of the class I major histocompatibility complex. Therefore, it is possible that these intracellular peptides play a physiological role.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Peptides/chemistry , Proteomics , beta 2-Microglobulin/chemistry , ATP Binding Cassette Transporter, Subfamily B, Member 2 , ATP-Binding Cassette Transporters/genetics , Amino Acid Sequence , Animals , Chromatography, Liquid , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Tandem Mass Spectrometry , beta 2-Microglobulin/genetics
6.
J Biol Chem ; 284(21): 14105-16, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19282285

ABSTRACT

Thimet oligopeptidase (EC 3.4.24.15; EP24.15) is an intracellular enzyme that has been proposed to metabolize peptides within cells, thereby affecting antigen presentation and G protein-coupled receptor signal transduction. However, only a small number of intracellular substrates of EP24.15 have been reported previously. Here we have identified over 100 peptides in human embryonic kidney 293 (HEK293) cells that are derived from intracellular proteins; many but not all of these peptides are substrates or products of EP24.15. First, cellular peptides were extracted from HEK293 cells and incubated in vitro with purified EP24.15. Then the peptides were labeled with isotopic tags and analyzed by mass spectrometry to obtain quantitative data on the extent of cleavage. A related series of experiments tested the effect of overexpression of EP24.15 on the cellular levels of peptides in HEK293 cells. Finally, synthetic peptides that corresponded to 10 of the cellular peptides were incubated with purified EP24.15 in vitro, and the cleavage was monitored by high pressure liquid chromatography and mass spectrometry. Many of the EP24.15 substrates identified by these approaches are 9-11 amino acids in length, supporting the proposal that EP24.15 can function in the degradation of peptides that could be used for antigen presentation. However, EP24.15 also converts some peptides into products that are 8-10 amino acids, thus contributing to the formation of peptides for antigen presentation. In addition, the intracellular peptides described here are potential candidates to regulate protein interactions within cells.


Subject(s)
Intracellular Space/enzymology , Metalloendopeptidases/metabolism , Amino Acid Sequence , Animals , Cell Extracts , Cell Line , Humans , Intracellular Space/drug effects , Isotope Labeling , Molecular Sequence Data , Peptides/chemistry , Quaternary Ammonium Compounds/pharmacology , Rats , Sequence Analysis, Protein , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity/drug effects
7.
J Biol Chem ; 283(36): 24448-59, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18617518

ABSTRACT

Protein degradation by the ubiquitin proteasome system releases large amounts of oligopeptides within cells. To investigate possible functions for these intracellularly generated oligopeptides, we fused them to a cationic transactivator peptide sequence using reversible disulfide bonds, introduced them into cells, and analyzed their effect on G protein-coupled receptor (GPCR) signal transduction. A mixture containing four of these peptides (20-80 microm) significantly inhibited the increase in the extracellular acidification response triggered by angiotensin II (ang II) in CHO-S cells transfected with the ang II type 1 receptor (AT1R-CHO-S). Subsequently, either alone or in a mixture, these peptides increased luciferase gene transcription in AT1R CHO-S cells stimulated with ang II and in HEK293 cells treated with isoproterenol. These peptides without transactivator failed to affect GPCR cellular responses. All four functional peptides were shown in vitro to competitively inhibit the degradation of a synthetic substrate by thimet oligopeptidase. Overexpression of thimet oligopeptidase in both CHO-S and HEK293 cells was sufficient to reduce luciferase activation triggered by a specific GPCR agonist. Moreover, using individual peptides as baits in affinity columns, several proteins involved in GPCR signaling were identified, including alpha-adaptin A and dynamin 1. These results suggest that before their complete degradation, intracellular peptides similar to those generated by proteasomes can actively affect cell signaling, probably representing additional bioactive molecules within cells.


Subject(s)
Oligopeptides/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology , Adaptor Protein Complex alpha Subunits/genetics , Adaptor Protein Complex alpha Subunits/metabolism , Angiotensin II/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Dynamin I/genetics , Dynamin I/metabolism , Gene Expression , Humans , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/genetics , Oligopeptides/genetics , Proteasome Endopeptidase Complex/genetics , Rats , Receptor, Angiotensin, Type 1/genetics , Signal Transduction/drug effects , Transfection , Ubiquitin/genetics , Ubiquitin/metabolism , Vasoconstrictor Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...