Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
STAR Protoc ; 1(2): 100067, 2020 09 18.
Article in English | MEDLINE | ID: mdl-33111105

ABSTRACT

Stimulation of naive T lymphocytes via the T cell receptor (TCR) induces distinct phosphorylation patterns that can be used to explore various signaling pathways within the cell. This protocol can be used to characterize different perturbations to the signaling pathways and the variations in time of stimulation. Here, we provide a method of barcoding and consolidating a maximum of 24 different sample conditions using two florescent dyes. This single sample for phospho-staining and flow cytometry saves time and reagents. For complete details on the use and execution of this protocol, please refer to Krutzik and Nolan (2006), Krutzik et al. (2012), Vercoulen et al. (2017), Ksionda et al. (2018), and Myers et al. (2019).


Subject(s)
Flow Cytometry/methods , Fluorescent Dyes , Molecular Probe Techniques , T-Lymphocytes , Animals , Cells, Cultured , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Mice , Phosphorylation , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Time Factors
2.
Oncogene ; 39(45): 6920-6934, 2020 11.
Article in English | MEDLINE | ID: mdl-32989257

ABSTRACT

Oncogenic mutations in RAS genes, like KRASG12D or NRASG12D, trap Ras in the active state and cause myeloproliferative disorder and T cell leukemia (T-ALL) when induced in the bone marrow via Mx1CRE. The RAS exchange factor RASGRP1 is frequently overexpressed in T-ALL patients. In T-ALL cell lines overexpression of RASGRP1 increases flux through the RASGTP/RasGDP cycle. Here we expanded RASGRP1 expression surveys in pediatric T-ALL and generated a RoLoRiG mouse model crossed to Mx1CRE to determine the consequences of induced RASGRP1 overexpression in primary hematopoietic cells. RASGRP1-overexpressing, GFP-positive cells outcompeted wild type cells and dominated the peripheral blood compartment over time. RASGRP1 overexpression bestows gain-of-function colony formation properties to bone marrow progenitors in medium containing limited growth factors. RASGRP1 overexpression enhances baseline mTOR-S6 signaling in the bone marrow, but not in vitro cytokine-induced signals. In agreement with these mechanistic findings, hRASGRP1-ires-EGFP enhances fitness of stem- and progenitor- cells, but only in the context of native hematopoiesis. RASGRP1 overexpression is distinct from KRASG12D or NRASG12D, does not cause acute leukemia on its own, and leukemia virus insertion frequencies predict that RASGRP1 overexpression can effectively cooperate with lesions in many other genes to cause acute T-ALL.


Subject(s)
DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Animals , Bone Marrow/pathology , Cells, Cultured , Child , Colony-Forming Units Assay , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Leukemic , Guanine Nucleotide Exchange Factors/genetics , Hematopoietic Stem Cell Transplantation , Humans , Male , Mice , Mice, Transgenic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Primary Cell Culture , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Transplantation Chimera
3.
Cell Rep ; 27(2): 631-647.e5, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30970263

ABSTRACT

Deregulated signal transduction is a cancer hallmark, and its complexity and interconnectivity imply that combination therapy should be considered, but large data volumes that cover the complexity are required in user-friendly ways. Here, we present a searchable database resource of synthetic lethality with a PI3 kinase signal transduction inhibitor by performing a saturation screen with an ultra-complex shRNA library containing 30 independent shRNAs per gene target. We focus on Ras-PI3 kinase signaling with T cell leukemia as a screening platform for multiple clinical and experimental reasons. Our resource predicts multiple combination-based therapies with high fidelity, ten of which we confirmed with small molecule inhibitors. Included are biochemical assays, as well as the IPI145 (duvelisib) inhibitor. We uncover the mechanism of synergy between the PI3 kinase inhibitor GDC0941 (pictilisib) and the tubulin inhibitor vincristine and demonstrate broad synergy in 28 cell lines of 5 cancer types and efficacy in preclinical leukemia mouse trials.


Subject(s)
Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , RNA, Small Interfering/genetics , Synthetic Lethal Mutations/genetics , Animals , Disease Models, Animal , Humans , Mice , Protein Kinase Inhibitors/pharmacology , Signal Transduction
4.
PLoS One ; 13(5): e0193849, 2018.
Article in English | MEDLINE | ID: mdl-29799846

ABSTRACT

T cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic cancer. Poly-chemotherapy with cytotoxic and genotoxic drugs causes substantial toxicity and more specific therapies targeting the underlying molecular lesions are highly desired. Perturbed Ras signaling is prevalent in T-ALL and occurs via oncogenic RAS mutations or through overexpression of the Ras activator RasGRP1 in ~65% of T-ALL patients. Effective small molecule inhibitors for either target do not currently exist. Genetic and biochemical evidence link phosphoinositide 3-kinase (PI3K) signals to T-ALL, PI3Ks are activated by Ras-dependent and Ras-independent mechanisms, and potent PI3K inhibitors exist. Here we performed comprehensive analyses of PI3K-Akt signaling in T-ALL with a focus on class I PI3K. We developed a multiplex, multiparameter flow cytometry platform with pan- and isoform-specific PI3K inhibitors. We find that pan-PI3K and PI3K γ-specific inhibitors effectively block basal and cytokine-induced PI3K-Akt signals. Despite such inhibition, GDC0941 (pan-PI3K) or AS-605240 (PI3Kγ-specific) as single agents did not efficiently induce death in T-ALL cell lines. Combination of GDC0941 with AS-605240, maximally targeting all p110 isoforms, exhibited potent synergistic activity for clonal T-ALL lines in vitro, which motivated us to perform preclinical trials in mice. In contrast to clonal T-ALL lines, we used a T-ALL cancer model that recapitulates the multi-step pathogenesis and inter- and intra-tumoral genetic heterogeneity, a hallmark of advanced human cancers. We found that the combination of GDC0941 with AS-605240 fails in such trials. Our results reveal that PI3K inhibitors are a promising avenue for molecular therapy in T-ALL, but predict the requirement for methods that can resolve biochemical signals in heterogeneous cell populations so that combination therapy can be designed in a rational manner.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Phosphoinositide-3 Kinase Inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Humans , Indazoles/pharmacology , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-akt/genetics , Sulfonamides/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Immunol ; 195(1): 31-5, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26002977

ABSTRACT

Ras GTPase-activating proteins (RasGAPs) inhibit signal transduction initiated through the Ras small GTP-binding protein. However, which members of the RasGAP family act as negative regulators of T cell responses is not completely understood. In this study, we investigated potential roles for the RasGAPs RASA1 and neurofibromin 1 (NF1) in T cells through the generation and analysis of T cell-specific RASA1 and NF1 double-deficient mice. In contrast to mice lacking either RasGAP alone in T cells, double-deficient mice developed T cell acute lymphoblastic leukemia/lymphoma, which originated at an early point in T cell development and was dependent on activating mutations in the Notch1 gene. These findings highlight RASA1 and NF1 as cotumor suppressors in the T cell lineage.


Subject(s)
Neurofibromin 1/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptor, Notch1/genetics , p120 GTPase Activating Protein/genetics , Animals , Gene Deletion , Gene Expression Regulation , Mice , Mice, Knockout , Mutation , Neurofibromin 1/deficiency , Neurofibromin 1/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/immunology , Signal Transduction , Spleen/immunology , Spleen/pathology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thymus Gland/immunology , Thymus Gland/pathology , Time Factors , p120 GTPase Activating Protein/deficiency , p120 GTPase Activating Protein/immunology
6.
J Immunol ; 192(6): 2830-2836, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24532586

ABSTRACT

Macrophages are centrally involved in the pathogenesis of acute inflammatory diseases, peritonitis, endotoxemia, and septic shock. However, the molecular mechanisms controlling such macrophage activation are incompletely understood. In this article, we provide evidence that Vav1, a member of the RhoGEF family, plays a crucial role in macrophage activation and septic endotoxemia. Vav1-deficient mice demonstrated a significantly increased susceptibility for LPS endotoxemia that could be abrogated by anti-IL-6R Ab treatment. Subsequent studies showed that Vav1-deficient macrophages display augmented production of the proinflammatory cytokine IL-6. Nuclear Vav1 was identified as a key negative regulator of macrophage-derived IL-6 production. In fact, Vav1 formed a nuclear DNA-binding complex with heat shock transcription factor 1 at the HSE2 region of the IL-6 promoter to suppress IL-6 gene transcription in macrophages. These findings provide new insights into the pathogenesis of endotoxemia and suggest new avenues for therapy.


Subject(s)
Endotoxemia/immunology , Interleukin-6/immunology , Macrophages/immunology , Proto-Oncogene Proteins c-vav/immunology , Animals , Blotting, Western , Cells, Cultured , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Endotoxemia/chemically induced , Endotoxemia/genetics , Gene Expression/immunology , Heat Shock Transcription Factors , Interleukin-10/blood , Interleukin-10/immunology , Interleukin-6/blood , Interleukin-6/genetics , Kaplan-Meier Estimate , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Proto-Oncogene Proteins c-vav/deficiency , Proto-Oncogene Proteins c-vav/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/immunology , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
7.
Sci Signal ; 6(268): ra21, 2013 Mar 26.
Article in English | MEDLINE | ID: mdl-23532335

ABSTRACT

Enhanced signaling by the small guanosine triphosphatase Ras is common in T cell acute lymphoblastic leukemia/lymphoma (T-ALL), but the underlying mechanisms are unclear. We identified the guanine nucleotide exchange factor RasGRP1 (Rasgrp1 in mice) as a Ras activator that contributes to leukemogenesis. We found increased RasGRP1 expression in many pediatric T-ALL patients, which is not observed in rare early T cell precursor T-ALL patients with KRAS and NRAS mutations, such as K-Ras(G12D). Leukemia screens in wild-type mice, but not in mice expressing the mutant K-Ras(G12D) that encodes a constitutively active Ras, yielded frequent retroviral insertions that led to increased Rasgrp1 expression. Rasgrp1 and oncogenic K-Ras(G12D) promoted T-ALL through distinct mechanisms. In K-Ras(G12D) T-ALLs, enhanced Ras activation had to be uncoupled from cell cycle arrest to promote cell proliferation. In mouse T-ALL cells with increased Rasgrp1 expression, we found that Rasgrp1 contributed to a previously uncharacterized cytokine receptor-activated Ras pathway that stimulated the proliferation of T-ALL cells in vivo, which was accompanied by dynamic patterns of activation of effector kinases downstream of Ras in individual T-ALLs. Reduction of Rasgrp1 abundance reduced cytokine-stimulated Ras signaling and decreased the proliferation of T-ALL in vivo. The position of RasGRP1 downstream of cytokine receptors as well as the different clinical outcomes that we observed as a function of RasGRP1 abundance make RasGRP1 an attractive future stratification marker for T-ALL.


Subject(s)
Enzyme Activation/physiology , Gene Expression Regulation, Neoplastic/physiology , Guanine Nucleotide Exchange Factors/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Signal Transduction/physiology , ras Proteins/metabolism , Animals , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Proliferation , Child , DNA Primers/genetics , Diglycerides , Gene Expression Profiling , Humans , Mice , Models, Biological , Mutagenesis, Insertional , Oligonucleotides/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
8.
Front Biol (Beijing) ; 8(5): 508-532, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24744772

ABSTRACT

RasGRP proteins are activators of Ras and other related small GTPases by the virtue of functioning as guanine nucleotide exchange factors (GEFs). In vertebrates, four RasGRP family members have been described. RasGRP-1 through -4 share many structural domains but there are also subtle differences between each of the different family members. Whereas SOS RasGEFs are ubiquitously expressed, RasGRP proteins are expressed in distinct patterns, such as in different cells of the hematopoietic system and in the brain. Most studies have concentrated on the role of RasGRP proteins in the development and function of immune cell types because of the predominant RasGRP expression profiles in these cells and the immune phenotypes of mice deficient for Rasgrp genes. However, more recent studies demonstrate that RasGRPs also play an important role in tumorigenesis. Examples are skin- and hematological-cancers but also solid malignancies such as melanoma or prostate cancer. These novel studies bring up many new and unanswered questions related to the molecular mechanism of RasGRP-driven oncogenesis, such as new receptor systems that RasGRP appears to respond to as well as regulatory mechanism for RasGRP expression that appear to be perturbed in these cancers. Here we will review some of the known aspects of RasGRP biology in lymphocytes and will discuss the exciting new notion that RasGRP Ras exchange factors play a role in oncogenesis downstream of various growth factor receptors.

9.
J Cell Sci ; 125(Pt 22): 5302-14, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22956543

ABSTRACT

The antigen-specific binding of T cells to antigen presenting cells results in recruitment of signalling proteins to microclusters at the cell-cell interface known as the immunological synapse (IS). The Vav1 guanine nucleotide exchange factor plays a critical role in T cell antigen receptor (TCR) signalling, leading to the activation of multiple pathways. We now show that it is recruited to microclusters and to the IS in primary CD4(+) and CD8(+) T cells. Furthermore, we show that this recruitment depends on the SH2 and C-terminal SH3 (SH3(B)) domains of Vav1, and on phosphotyrosines 112 and 128 of the SLP76 adaptor protein. Biophysical measurements show that Vav1 binds directly to these residues on SLP76 and that efficient binding depends on the SH2 and SH3(B) domains of Vav1. Finally, we show that the same two domains are critical for the phosphorylation of Vav1 and its signalling function in TCR-induced calcium flux. We propose that Vav1 is recruited to the IS by binding to SLP76 and that this interaction is critical for the transduction of signals leading to calcium flux.


Subject(s)
Proto-Oncogene Proteins c-vav/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Cells, Cultured , Humans , Immunological Synapses/metabolism , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phosphorylation , Protein Transport/immunology , Proto-Oncogene Proteins c-vav/chemistry , src Homology Domains
10.
Sci Signal ; 4(193): ra66, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21971040

ABSTRACT

During T cell activation by antigen-presenting cells (APCs), the diverse spatiotemporal organization of components of T cell signaling pathways modulates the efficiency of activation. Here, we found that loss of the tyrosine kinase interleukin-2 (IL-2)-inducible T cell kinase (Itk) in mice altered the spatiotemporal distributions of 14 of 16 sensors of T cell signaling molecules in the region of the interface between the T cell and the APC, which reduced the segregation of signaling intermediates into distinct spatiotemporal patterns. Activation of the Rho family guanosine triphosphatase Cdc42 at the center of the cell-cell interface was impaired, although the total cellular amount of active Cdc42 remained intact. The defect in Cdc42 localization resulted in impaired actin accumulation at the T cell-APC interface in Itk-deficient T cells. Reconstitution of cells with active Cdc42 that was specifically directed to the center of the interface restored actin accumulation in Itk-deficient T cells. Itk also controlled the central localization of the guanine nucleotide exchange factor SLAT [Switch-associated protein 70 (SWAP-70)-like adaptor of T cells], which may contribute to the activation of Cdc42 at the center of the interface. Together, these data illustrate how control of the spatiotemporal organization of T cell signaling controls critical aspects of T cell function.


Subject(s)
Lymphocyte Activation/immunology , Protein-Tyrosine Kinases/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Actins/genetics , Actins/immunology , Actins/metabolism , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Enzyme Activation/genetics , Enzyme Activation/immunology , Guanine Nucleotide Exchange Factors , Lymphocyte Activation/genetics , Mice , Mice, Transgenic , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Signal Transduction/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/immunology , cdc42 GTP-Binding Protein/metabolism
11.
Sci Signal ; 2(101): ra83, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-20009105

ABSTRACT

The guanine nucleotide exchange factor (GEF) Vav1 is essential for transducing T cell antigen receptor (TCR) signals and therefore plays a critical role in the development and activation of T cells. It has been presumed that the GEF activity of Vav1 is important for its function; however, there has been no direct demonstration of this. Here, we generated mice expressing enzymatically inactive, but normally folded, Vav1 protein. Analysis of these mice showed that the GEF activity of Vav1 was necessary for the selection of thymocytes and for the optimal activation of T cells, including signal transduction to Rac1, Akt, and integrins. In contrast, the GEF activity of Vav1 was not required for TCR-induced calcium flux, activation of extracellular signal-regulated kinase and protein kinase D1, and cell polarization. Thus, in T cells, the GEF activity of Vav1 is essential for some, but not all, of its functions.


Subject(s)
Guanine Nucleotide Exchange Factors/physiology , Proto-Oncogene Proteins c-vav/physiology , T-Lymphocytes/cytology , Animals , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins c-vav/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...