Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Commun Signal ; 22(1): 332, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38886790

ABSTRACT

BACKGROUND: HRASKO/NRASKO double knockout mice exhibit exceedingly high rates of perinatal lethality due to respiratory failure caused by a significant lung maturation delay. The few animals that reach adulthood have a normal lifespan, but present areas of atelectasis mixed with patches of emphysema and normal tissue in the lung. METHODS: Eight double knockout and eight control mice were analyzed using micro-X-ray computerized tomography and a Small Animal Physiological Monitoring system. Tissues and samples from these mice were analyzed using standard histological and Molecular Biology methods and the significance of the results analyzed using a Student´s T-test. RESULTS: The very few double knockout mice surviving up to adulthood display clear craniofacial abnormalities reminiscent of those seen in RASopathy mouse models, as well as thrombocytopenia, bleeding anomalies, and reduced platelet activation induced by thrombin. These surviving mice also present heart and spleen hyperplasia, and elevated numbers of myeloid-derived suppressor cells in the spleen. Mechanistically, we observed that these phenotypic alterations are accompanied by increased KRAS-GTP levels in heart, platelets and primary mouse embryonic fibroblasts from these animals. CONCLUSIONS: Our data uncovers a new, previously unidentified mechanism capable of triggering a RASopathy phenotype in mice as a result of the combined removal of HRAS and NRAS.


Subject(s)
GTP Phosphohydrolases , Mice, Knockout , Phenotype , Proto-Oncogene Proteins p21(ras) , Animals , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Mice , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Platelet Activation/genetics , Spleen/pathology , Spleen/metabolism , Monomeric GTP-Binding Proteins
2.
Nat Commun ; 14(1): 5856, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37730692

ABSTRACT

The impact of genetic ablation of SOS1 or SOS2 is evaluated in a murine model of KRASG12D-driven lung adenocarcinoma (LUAD). SOS2 ablation shows some protection during early stages but only SOS1 ablation causes significant, specific long term increase of survival/lifespan of the KRASG12D mice associated to markedly reduced tumor burden and reduced populations of cancer-associated fibroblasts, macrophages and T-lymphocytes in the lung tumor microenvironment (TME). SOS1 ablation also causes specific shrinkage and regression of LUAD tumoral masses and components of the TME in pre-established KRASG12D LUAD tumors. The critical requirement of SOS1 for KRASG12D-driven LUAD is further confirmed by means of intravenous tail injection of KRASG12D tumor cells into SOS1KO/KRASWT mice, or of SOS1-less, KRASG12D tumor cells into wildtype mice. In silico analyses of human lung cancer databases support also the dominant role of SOS1 regarding tumor development and survival in LUAD patients. Our data indicate that SOS1 is critically required for development of KRASG12D-driven LUAD and confirm the validity of this RAS-GEF activator as an actionable therapeutic target in KRAS mutant LUAD.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Cancer-Associated Fibroblasts , Lung Neoplasms , Animals , Humans , Mice , Adenocarcinoma/genetics , Adenocarcinoma of Lung/genetics , Lung Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Microenvironment/genetics
3.
Oncogene ; 42(5): 389-405, 2023 01.
Article in English | MEDLINE | ID: mdl-36476833

ABSTRACT

The R-RAS2 GTP hydrolase (GTPase) (also known as TC21) has been traditionally considered quite similar to classical RAS proteins at the regulatory and signaling levels. Recently, a long-tail hotspot mutation targeting the R-RAS2/TC21 Gln72 residue (Q72L) was identified as a potent oncogenic driver. Additional point mutations were also found in other tumors at low frequencies. Despite this, little information is available regarding the transforming role of these mutant versions and their relevance for the tumorigenic properties of already-transformed cancer cells. Here, we report that many of the RRAS2 mutations found in human cancers are highly transforming when expressed in immortalized cell lines. Moreover, the expression of endogenous R-RAS2Q72L is important for maintaining optimal levels of PI3K and ERK activities as well as for the adhesion, invasiveness, proliferation, and mitochondrial respiration of ovarian and breast cancer cell lines. Endogenous R-RAS2Q72L also regulates gene expression programs linked to both cell adhesion and inflammatory/immune-related responses. Endogenous R-RAS2Q72L is also quite relevant for the in vivo tumorigenic activity of these cells. This dependency is observed even though these cancer cell lines bear concurrent gain-of-function mutations in genes encoding RAS signaling elements. Finally, we show that endogenous R-RAS2, unlike the case of classical RAS proteins, specifically localizes in focal adhesions. Collectively, these results indicate that gain-of-function mutations of R-RAS2/TC21 play roles in tumor initiation and maintenance that are not fully redundant with those regulated by classical RAS oncoproteins.


Subject(s)
Monomeric GTP-Binding Proteins , Neoplasms , Humans , Cell Line , Monomeric GTP-Binding Proteins/genetics , Neoplasms/genetics , ras Proteins/genetics , ras Proteins/metabolism , Signal Transduction/genetics
4.
Cancers (Basel) ; 14(16)2022 Aug 11.
Article in English | MEDLINE | ID: mdl-36010887

ABSTRACT

We showed previously that the ABL-mediated phosphorylation of SOS1 promotes RAC activation and contributes to BCR-ABL leukemogenesis, suggesting the relevant role of SOS1 in the pathogenesis of CML. To try and obtain direct experimental evidence of the specific mechanistic implication of SOS1 in CML development, here, we combined a murine model of CML driven by a p210BCR/ABL transgene with our tamoxifen-inducible SOS1/2-KO system in order to investigate the phenotypic impact of the direct genetic ablation of SOS1 or SOS2 on the pathogenesis of CML. Our observations showed that, in contrast to control animals expressing normal levels of SOS1 and SOS2 or to single SOS2-KO mice, p210BCR/ABL transgenic mice devoid of SOS1 presented significantly extended survival curves and also displayed an almost complete disappearance of the typical hematological alterations and splenomegaly constituting the hallmarks of CML. SOS1 ablation also resulted in a specific reduction in the proliferation and the total number of colony-forming units arising from the population of bone marrow stem/progenitor cells from p210BCR/ABL transgenic mice. The specific blockade of CML development caused by SOS1 ablation in p210BCR/ABL mice indicates that SOS1 is critically required for CML pathogenesis and supports the consideration of this cellular GEF as a novel, alternative bona fide therapeutic target for CML treatment in the clinic.

5.
Biomolecules ; 11(8)2021 07 30.
Article in English | MEDLINE | ID: mdl-34439794

ABSTRACT

Recent breakthroughs have reignited interest in RAS GEFs as direct therapeutic targets. To search for new inhibitors of SOS GEF activity, a repository of known/approved compounds (NIH-NACTS) and a library of new marine compounds (Biomar Microbial Technologies) were screened by means of in vitro RAS-GEF assays using purified, bacterially expressed SOS and RAS constructs. Interestingly, all inhibitors identified in our screenings (two per library) shared related chemical structures belonging to the anthraquinone family of compounds. All our anthraquinone SOS inhibitors were active against the three canonical RAS isoforms when tested in our SOS GEF assays, inhibited RAS activation in mouse embryonic fibroblasts, and were also able to inhibit the growth of different cancer cell lines harboring WT or mutant RAS genes. In contrast to the commercially available anthraquinone inhibitors, our new marine anthraquinone inhibitors did not show in vivo cardiotoxicity, thus providing a lead for future discovery of stronger, clinically useful anthraquinone SOS GEF blockers.


Subject(s)
Anthraquinones/pharmacology , Antineoplastic Agents/pharmacology , GTP Phosphohydrolases/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Cardiotoxicity/prevention & control , Cell Line, Transformed , Cell Line, Tumor , Doxorubicin/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Idarubicin/pharmacology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Knockout , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , SOS1 Protein/genetics , SOS1 Protein/metabolism , Son of Sevenless Proteins/deficiency , Son of Sevenless Proteins/genetics
6.
Int J Mol Sci ; 22(12)2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34205562

ABSTRACT

The SOS family of Ras-GEFs encompasses two highly homologous and widely expressed members, SOS1 and SOS2. Despite their similar structures and expression patterns, early studies of constitutive KO mice showing that SOS1-KO mutants were embryonic lethal while SOS2-KO mice were viable led to initially viewing SOS1 as the main Ras-GEF linking external stimuli to downstream RAS signaling, while obviating the functional significance of SOS2. Subsequently, different genetic and/or pharmacological ablation tools defined more precisely the functional specificity/redundancy of the SOS1/2 GEFs. Interestingly, the defective phenotypes observed in concomitantly ablated SOS1/2-DKO contexts are frequently much stronger than in single SOS1-KO scenarios and undetectable in single SOS2-KO cells, demonstrating functional redundancy between them and suggesting an ancillary role of SOS2 in the absence of SOS1. Preferential SOS1 role was also demonstrated in different RASopathies and tumors. Conversely, specific SOS2 functions, including a critical role in regulation of the RAS-PI3K/AKT signaling axis in keratinocytes and KRAS-driven tumor lines or in control of epidermal stem cell homeostasis, were also reported. Specific SOS2 mutations were also identified in some RASopathies and cancer forms. The relevance/specificity of the newly uncovered functional roles suggests that SOS2 should join SOS1 for consideration as a relevant biomarker/therapy target.


Subject(s)
SOS1 Protein/physiology , Son of Sevenless Proteins/physiology , Animals , Humans , Neoplasms/metabolism
7.
Sci Rep ; 11(1): 12574, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34131176

ABSTRACT

Human neutrophils constitutively express high amounts of arginase-1, which depletes arginine from the surrounding medium and downregulates T-cell activation. Here, we have found that neutrophil arginase-1, released from activated human neutrophils or dead cells, induced apoptosis in cancer cells through an endoplasmic reticulum (ER) stress pathway. Silencing of PERK in cancer cells prevented the induction of ER stress and apoptosis. Arginase inhibitor Nω-hydroxy-nor-arginine inhibited apoptosis and ER stress response induced by conditioned medium from activated neutrophils. A number of tumor cell lines, derived from different tissues, were sensitive to neutrophil arginase-1, with pancreatic, breast, ovarian and lung cancer cells showing the highest sensitivity. Neutrophil-released arginase-1 and arginine deprivation potentiated the antitumor action against pancreatic cancer cells of the ER-targeted antitumor alkylphospholipid analog edelfosine. Our study demonstrates the involvement of neutrophil arginase-1 in cancer cell killing and highlights the importance and complex role of neutrophils in tumor surveillance and biology.


Subject(s)
Arginase/genetics , Endoplasmic Reticulum Stress/genetics , T-Lymphocytes/immunology , eIF-2 Kinase/genetics , Apoptosis/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Endoplasmic Reticulum/enzymology , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Neutrophils/enzymology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , T-Lymphocytes/enzymology
8.
Oncogene ; 40(27): 4538-4551, 2021 07.
Article in English | MEDLINE | ID: mdl-34120142

ABSTRACT

SOS1 ablation causes specific defective phenotypes in MEFs including increased levels of intracellular ROS. We showed that the mitochondria-targeted antioxidant MitoTEMPO restores normal endogenous ROS levels, suggesting predominant involvement of mitochondria in generation of this defective SOS1-dependent phenotype. The absence of SOS1 caused specific alterations of mitochondrial shape, mass, and dynamics accompanied by higher percentage of dysfunctional mitochondria and lower rates of electron transport in comparison to WT or SOS2-KO counterparts. SOS1-deficient MEFs also exhibited specific alterations of respiratory complexes and their assembly into mitochondrial supercomplexes and consistently reduced rates of respiration, glycolysis, and ATP production, together with distinctive patterns of substrate preference for oxidative energy metabolism and dependence on glucose for survival. RASless cells showed defective respiratory/metabolic phenotypes reminiscent of those of SOS1-deficient MEFs, suggesting that the mitochondrial defects of these cells are mechanistically linked to the absence of SOS1-GEF activity on cellular RAS targets. Our observations provide a direct mechanistic link between SOS1 and control of cellular oxidative stress and suggest that SOS1-mediated RAS activation is required for correct mitochondrial dynamics and function.


Subject(s)
Mitochondrial Dynamics , Homeostasis , ras Guanine Nucleotide Exchange Factors
9.
Cancers (Basel) ; 13(9)2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33946974

ABSTRACT

Prior reports showed the critical requirement of Sos1 for epithelial carcinogenesis, but the specific functionalities of the homologous Sos1 and Sos2 GEFs in skin homeostasis and tumorigenesis remain unclear. Here, we characterize specific mechanistic roles played by Sos1 or Sos2 in primary mouse keratinocytes (a prevalent skin cell lineage) under different experimental conditions. Functional analyses of actively growing primary keratinocytes of relevant genotypes-WT, Sos1-KO, Sos2-KO, and Sos1/2-DKO-revealed a prevalent role of Sos1 regarding transcriptional regulation and control of RAS activation and mechanistic overlapping of Sos1 and Sos2 regarding cell proliferation and survival, with dominant contribution of Sos1 to the RAS-ERK axis and Sos2 to the RAS-PI3K/AKT axis. Sos1/2-DKO keratinocytes could not grow under 3D culture conditions, but single Sos1-KO and Sos2-KO keratinocytes were able to form pseudoepidermis structures that showed disorganized layer structure, reduced proliferation, and increased apoptosis in comparison with WT 3D cultures. Remarkably, analysis of the skin of both newborn and adult Sos2-KO mice uncovered a significant reduction of the population of stem cells located in hair follicles. These data confirm that Sos1 and Sos2 play specific, cell-autonomous functions in primary keratinocytes and reveal a novel, essential role of Sos2 in control of epidermal stem cell homeostasis.

10.
Mol Cell Biol ; 38(16)2018 08 15.
Article in English | MEDLINE | ID: mdl-29844066

ABSTRACT

Using Sos1 knockout (Sos1-KO), Sos2-KO, and Sos1/2 double-knockout (Sos1/2-DKO) mice, we assessed the functional role of Sos1 and Sos2 in skin homeostasis under physiological and/or pathological conditions. Sos1 depletion resulted in significant alterations of skin homeostasis, including reduced keratinocyte proliferation, altered hair follicle and blood vessel integrity in dermis, and reduced adipose tissue in hypodermis. These defects worsened significantly when both Sos1 and Sos2 were absent. Simultaneous Sos1/2 disruption led to severe impairment of the ability to repair skin wounds, as well as to almost complete ablation of the neutrophil-mediated inflammatory response in the injury site. Furthermore, Sos1 disruption delayed the onset of tumor initiation, decreased tumor growth, and prevented malignant progression of papillomas in a DMBA (7,12-dimethylbenz[α]anthracene)/TPA (12-O-tetradecanoylphorbol-13-acetate)-induced skin carcinogenesis model. Finally, Sos1 depletion in preexisting chemically induced papillomas resulted also in decreased tumor growth, probably linked to significantly reduced underlying keratinocyte proliferation. Our data unveil novel, distinctive mechanistic roles of Sos 1 and Sos2 in physiological control of skin homeostasis and wound repair, as well as in pathological development of chemically induced skin tumors. These observations underscore the essential role of Sos proteins in cellular proliferation and migration and support the consideration of these RasGEFs as potential biomarkers/therapy targets in Ras-driven epidermal tumors.


Subject(s)
SOS1 Protein/metabolism , Skin Neoplasms/etiology , Skin/metabolism , Son of Sevenless Proteins/metabolism , Animals , Carcinogenesis , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Homeostasis , Mice , Mice, Knockout , Neovascularization, Physiologic , Papilloma/metabolism , Papilloma/pathology , SOS1 Protein/deficiency , SOS1 Protein/genetics , Skin/blood supply , Skin/cytology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Son of Sevenless Proteins/deficiency , Son of Sevenless Proteins/genetics , Wound Healing
11.
Mol Cell Neurosci ; 85: 127-147, 2017 12.
Article in English | MEDLINE | ID: mdl-28966131

ABSTRACT

Various parameters of neurogenesis were analyzed in parallel in the two neurogenic areas (the Dentate Gyrus[DG] and the Subventricular Zone[SVZ]/Rostral Migratory Stream[RMS]/Main Olfactory Bulb[MOB] neurogenic system) of adult WT and KO mouse strains for the Ras-GRF1/2 genes (Ras-GRF1-KO, Ras-GRF2-KO, Ras-GRF1/2-DKO). Significantly reduced numbers of doublecortin[DCX]-positive cells were specifically observed in the DG, but not the SVZ/RMS/MOB neurogenic region, of Ras-GRF2-KO and Ras-GRF1/2-DKO mice indicating that this novel Ras-GRF2-dependent phenotype is spatially restricted to a specific neurogenic area. Consistent with a role of CREB as mediator of Ras-GRF2 function in neurogenesis, the density of p-CREB-positive cells was also specifically reduced in all neurogenic regions of Ras-GRF2-KO and DKO mice. Similar levels of early neurogenic proliferation markers (Ki67, BrdU) were observed in all different Ras-GRF genotypes analyzed but significantly elevated levels of nestin-immunolabel, particularly of undifferentiated, highly ramified, A-type nestin-positive neurons were specifically detected in the DG but not the SVZ/RMS/MOB of Ras-GRF2-KO and DKO mice. Together with assays of other neurogenic markers (GFAP, Sox2, Tuj1, NeuN), these observations suggest that the deficit of DCX/p-CREB-positive cells in the DG of Ras-GRF2-depleted mice does not involve impaired neuronal proliferation but rather delayed transition from the stem cell stage to the differentiation stages of the neurogenic process. This model is also supported by functional analyses of DG-derived neurosphere cultures and transcriptional characterization of the neurogenic areas of mice of all relevant Ras-GRF genotypes suggesting that the neurogenic role of Ras-GRF2 is exerted in a cell-autonomous manner through a specific transcriptional program.


Subject(s)
Cell Differentiation/physiology , Dentate Gyrus/cytology , Neural Stem Cells/cytology , Neurogenesis/physiology , ras-GRF1/metabolism , Animals , Dentate Gyrus/metabolism , Doublecortin Protein , Guanine Nucleotide-Releasing Factor 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nestin
14.
Dalton Trans ; 42(38): 13955-67, 2013 Oct 14.
Article in English | MEDLINE | ID: mdl-23925580

ABSTRACT

New ruthenium complexes containing enantiopure 2,6-bis[4'(R)-phenyloxazolin-2'-il-pyridine] ((R,R)-Ph-pybox), 2,6-bis[4'(S)-isopropyloxazolin-2'-il-pyridine] ((S,S)-(i)Pr-pybox) or 2,6-bis[4'(R)-isopropyloxazolin-2'-il-pyridine] ((R,R)-(i)Pr-pybox) and water soluble 1,3,5-triaza-7-phosphaadamantane (PTA) or N-substituted PTA phosphanes have been synthesized in high yields and fully characterized. The interactions of these compounds with plasmidic DNA and their cytotoxic activity against the human cervical cancer HeLa cell line are reported, pointing out for the first time the different behaviour of ruthenium enantiomers affecting the cell cycle in HeLa tumor cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Organometallic Compounds/chemical synthesis , Oxazoles/chemical synthesis , Pyridines/chemical synthesis , Ruthenium/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , DNA/chemistry , HeLa Cells , Humans , Magnetic Resonance Spectroscopy , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Oxazoles/chemistry , Oxazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Ruthenium/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spectroscopy, Fourier Transform Infrared , Stereoisomerism
15.
J Med Chem ; 56(7): 2813-27, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23470139

ABSTRACT

Colchicine site ligands with indole B rings are potent tubulin polymerization inhibitors. Structural modifications at the indole 3-position of 1-methyl-5-indolyl-based isocombretastatins (1,1-diarylethenes) and phenstatins endowed them with anchors for further derivatization and resulted in highly potent compounds. The substituted derivatives displayed potent cytotoxicity against several human cancer cell lines due to tubulin inhibition, as shown by cell cycle analysis, confocal microscopy, and tubulin polymerization inhibitory activity studies and promoted cell killing mediated by caspase-3 activation. Binding at the colchicine site was confirmed by means of fluorescence measurements of MTC displacement. Molecular modeling suggests that the tropolone-binding region of the colchicine site of tubulin can adapt to hosting small polar substituents. Isocombretastatins accepted substitutions better than phenstatins, and the highest potencies were achieved for the cyano and hydroxyiminomethyl substituents, with TPI values in the submicromolar range and cytotoxicities in the subnanomolar range. A 3,4,5-trimethoxyphenyl ring usually afforded more potent derivatives than a 2,3,4-trimethoxyphenyl ring.


Subject(s)
Indoles/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Cell Line, Tumor , Humans , Indoles/chemistry
16.
Autophagy ; 8(11): 1557-76, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22874569

ABSTRACT

L-arginine (L-Arg) deficiency results in decreased T-cell proliferation and impaired T-cell function. Here we have found that L-Arg depletion inhibited expression of different membrane antigens, including CD247 (CD3ζ), and led to an ER stress response, as well as cell cycle arrest at G(0)/G(1) in both human Jurkat and peripheral blood mitogen-activated T cells, without undergoing apoptosis. By genetic and biochemical approaches, we found that L-Arg depletion also induced autophagy. Deprivation of L-Arg induced EIF2S1 (eIF2α), MAPK8 (JNK), BCL2 (Bcl-2) phosphorylation, and displacement of BECN1 (Beclin 1) binding to BCL2, leading to autophagosome formation. Silencing of ERN1 (IRE1α) prevented the induction of autophagy as well as MAPK8 activation, BCL2 phosphorylation and XBP1 splicing, whereas led T lymphocytes to apoptosis under L-Arg starvation, suggesting that the ERN1-MAPK8 pathway plays a major role in the activation of autophagy following L-Arg depletion. Autophagy was required for survival of T lymphocytes in the absence of L-Arg, and resulted in a reversible process. Replenishment of L-Arg made T lymphocytes to regain the normal cell cycle profile and proliferate, whereas autophagy was inhibited. Inhibition of autophagy by ERN1, BECN1 and ATG7 silencing, or by pharmacological inhibitors, promoted cell death of T lymphocytes incubated in the absence of L-Arg. Our data indicate for the first time that depletion of L-Arg in T lymphocytes leads to a reversible response that preserves T lymphocytes through ER stress and autophagy, while remaining arrested at G(0)/G(1). Our data also show that the L-Arg depletion-induced ER stress response could lead to apoptosis when autophagy is blocked.


Subject(s)
Arginine/deficiency , Arginine/pharmacology , Autophagy/drug effects , Cytoprotection/drug effects , Endoplasmic Reticulum Stress/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 7 , Beclin-1 , CD3 Complex/metabolism , Cell Proliferation/drug effects , DNA-Binding Proteins/genetics , Down-Regulation/drug effects , Endoribonucleases/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Jurkat Cells , Lymphocyte Activation/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Mitogens/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Splicing/drug effects , RNA Splicing/genetics , Regulatory Factor X Transcription Factors , T-Lymphocytes/enzymology , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/genetics , Ubiquitin-Activating Enzymes/metabolism , X-Box Binding Protein 1
17.
Eur J Immunol ; 41(12): 3436-42, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22229154

ABSTRACT

The P-selectin glycoprotein ligand-1 (PSGL-1) is involved in the initial contact of leukocytes with activated endothelium, and its adhesive function is regulated through its proteolytic processing. We have found that the metalloprotease ADAM8 is both associated with PSGL-1 through the ezrin­radixin­moesin actin-binding proteins and able to cause the proteolytic cleavage of this adhesion receptor. Accordingly, ADAM8 knockdown increases PSGL-1 expression, and functional assays show that ADAM8 is able to reduce leukocyte rolling on P-selectin and hence on activated endothelial cells. We conclude that ADAM8 modulates the expression and function of PSGL-1.


Subject(s)
ADAM Proteins/metabolism , DNA-Binding Proteins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Transcription Factors/metabolism , ADAM Proteins/genetics , Cell Line, Tumor , Cells, Cultured , Endothelium/metabolism , HL-60 Cells , Humans , Leukocyte Rolling/physiology , Leukocytes/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Microfilament Proteins/metabolism , Platelet Glycoprotein GPIb-IX Complex
18.
Dalton Trans ; 39(42): 10186-96, 2010 Nov 14.
Article in English | MEDLINE | ID: mdl-20882255

ABSTRACT

The synthesis and full characterization of new half-sandwich ruthenium(II) complexes containing κ(3)(N,N,N)-hydridotris(pyrazolyl)borate (κ(3)(N,N,N)-Tp) and the water-soluble phosphanes 1,3,5-triaza-7-phosphatricyclo[3.3.1.1(3,7)]decane (PTA) and 1-methyl-3,5-diaza-1-azonia-7-phosphatricyclo[3.3.1.1(3,7)]decane (1-CH(3)-PTA) has been explored. Single crystal X-ray diffraction analysis for complex [RuCl{κ(3)(N,N,N)-Tp}(PMe(2)Ph)(1-CH(3)-PTA)][CF(3)SO(3)]·2NCMe is also reported. DNA binding properties of the ruthenium complexes have been evaluated by mobility shift assay and MALDI-TOF mass spectrometry. The in vitro antitumor activity of these compounds was assessed by examining their ability to inhibit cell proliferation in a number of human cancer cell lines (NCI-H460, SF-268, MCF-7) and non-tumor human umbilical vein endothelial cells (HUVEC). Some of these new compounds show promising cytotoxic activity with IC(50) values in the low micromolar range, and display differential effects on cancer and normal cell growth.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Phosphines/chemistry , Pyrazoles/chemistry , Ruthenium/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/genetics , DNA/metabolism , Electrochemistry , Humans , Organometallic Compounds/chemical synthesis , Organometallic Compounds/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...