Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Hepatol ; 74(4): 893-906, 2021 04.
Article in English | MEDLINE | ID: mdl-33197513

ABSTRACT

BACKGROUND & AIMS: The tumour microenvironment shapes tumour growth through cellular communications that include both direct interactions and secreted factors. The aim of this study was to characterize the impact of the secreted glycoprotein ADAMTSL5, whose role in cancer has not been previously investigated, on hepatocellular carcinoma (HCC). METHODS: ADAMTSL5 methylation status was evaluated through bisulfite sequencing, and publicly available data analysis. ADAMTSL5 RNA and protein expression were assessed in mouse models and HCC patient samples and compared to data from published datasets. Functional studies, including association of ADAMTSL5 depletion with responsiveness to clinically relevant drugs, were performed in cellular and in vivo models. Molecular alterations associated with ADAMTSL5 targeting were determined using proteomics, biochemistry, and reverse-transcription quantitative PCR. RESULTS: Methylome analysis revealed hypermethylated gene body CpG islands at the ADAMTSL5 locus in both mouse and human HCC, correlating with higher ADAMTSL5 expression. ADAMTSL5 targeting interfered with tumorigenic properties of HCC cells in vitro and in vivo, whereas ADAMTSL5 overexpression conferred tumorigenicity to pre-tumoural hepatocytes sensitized to transformation by a modest level of MET receptor expression. Mechanistically, ADAMTSL5 abrogation led to a reduction of several oncogenic inputs relevant to HCC, including reduced expression and/or phosphorylation levels of receptor tyrosine kinases MET, EGFR, PDGFRß, IGF1Rß, or FGFR4. This phenotype was associated with significantly increased sensitivity of HCC cells to clinically relevant drugs, namely sorafenib, lenvatinib, and regorafenib. Moreover, ADAMTSL5 depletion drastically increased expression of AXL, accompanied by a sensitization to bemcentinib. CONCLUSIONS: Our results point to a role for ADAMTSL5 in maintaining the function of key oncogenic signalling pathways, suggesting that it may act as a master regulator of tumorigenicity and drug resistance in HCC. LAY SUMMARY: The environment of cancer cells has profound effects on establishment, progression, and response of a tumour to treatment. Herein, we show that ADAMTSL5, a protein secreted by liver cancer cells and overlooked in cancer so far, is increased in this tumour type, is necessary for tumour formation and supports drug resistance. Adamtsl5 removal conferred sensitivity of liver cancer cells to drugs used in current treatment. This suggests ADAMTSL5 as a potential marker in liver cancer as well as a possible drug target.


Subject(s)
ADAMTS Proteins , ADAMTS5 Protein , Carcinogenesis , Carcinoma, Hepatocellular , Drug Resistance, Neoplasm/physiology , Liver Neoplasms , Signal Transduction , ADAMTS Proteins/genetics , ADAMTS Proteins/metabolism , ADAMTS5 Protein/genetics , ADAMTS5 Protein/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Benzocycloheptenes/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Epigenomics , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Phenylurea Compounds/pharmacology , Quinolines/pharmacology , Sorafenib/pharmacology , Transcriptional Activation , Triazoles/pharmacology , Tumor Microenvironment/physiology
2.
Cancers (Basel) ; 12(8)2020 Aug 14.
Article in English | MEDLINE | ID: mdl-32823931

ABSTRACT

The complexity of hepatocellular carcinoma (HCC) challenges the identification of disease-relevant signals. C3G, a guanine nucleotide exchange factor for Rap and other Ras proteins, plays a dual role in cancer acting as either a tumor suppressor or promoter depending on tumor type and stage. The potential relevance of C3G upregulation in HCC patients suggested by database analysis remains unknown. We have explored C3G function in HCC and the underlying mechanisms using public patient data and in vitro and in vivo human and mouse HCC models. We found that C3G is highly expressed in progenitor cells and neonatal hepatocytes, whilst being down-regulated in adult hepatocytes and re-expressed in human HCC patients, mouse HCC models and HCC cell lines. Moreover, high C3G mRNA levels correlate with tumor progression and a lower patient survival rate. C3G expression appears to be tightly modulated within the HCC program, influencing distinct cell biological properties. Hence, high C3G expression levels are necessary for cell tumorigenic properties, as illustrated by reduced colony formation in anchorage-dependent and -independent growth assays induced by permanent C3G silencing using shRNAs. Additionally, we demonstrate that C3G down-regulation interferes with primary HCC tumor formation in xenograft assays, increasing apoptosis and decreasing proliferation. In vitro assays also revealed that C3G down-regulation enhances the pro-migratory, invasive and metastatic properties of HCC cells through an epithelial-mesenchymal switch that favors the acquisition of a more mesenchymal phenotype. Consistently, a low C3G expression in HCC cells correlates with lung metastasis formation in mice. However, the subsequent restoration of C3G levels is associated with metastatic growth. Mechanistically, C3G down-regulation severely impairs HGF/MET signaling activation in HCC cells. Collectively, our results indicate that C3G is a key player in HCC. C3G promotes tumor growth and progression, and the modulation of its levels is essential to ensure distinct biological features of HCC cells throughout the oncogenic program. Furthermore, C3G requirement for HGF/MET signaling full activation provides mechanistic data on how it works, pointing out the relevance of assessing whether high C3G levels could identify HCC responders to MET inhibitors.

3.
iScience ; 21: 68-83, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31655257

ABSTRACT

Computed tomography is a powerful medical imaging modality for longitudinal studies in cancer to follow neoplasia progression and evaluate anticancer therapies. Here, we report the generation of a photon-counting micro-computed tomography (PC-CT) method based on hybrid pixel detectors with enhanced sensitivity and precision of tumor imaging. We then applied PC-CT for longitudinal imaging in a clinically relevant liver cancer model, the Alb-R26Met mice, and found a remarkable heterogeneity in the dynamics for tumors at the initiation phases. Instead, the growth curve of evolving tumors exhibited a comparable exponential growth, with a constant doubling time. Furthermore, longitudinal PC-CT imaging in mice treated with a combination of MEK and BCL-XL inhibitors revealed a drastic tumor regression accompanied by a striking remodeling of macrophages in the tumor microenvironment. Thus, PC-CT is a powerful system to detect cancer initiation and progression, and to monitor its evolution during treatment.

4.
J Hepatol ; 70(3): 470-482, 2019 03.
Article in English | MEDLINE | ID: mdl-30529386

ABSTRACT

BACKGROUND & AIMS: The variety of alterations found in hepatocellular carcinoma (HCC) makes the identification of functionally relevant genes and their combinatorial actions in tumorigenesis challenging. Deregulation of receptor tyrosine kinases (RTKs) is frequent in HCC, yet little is known about the molecular events that cooperate with RTKs and whether these cooperative events play an active role at the root of liver tumorigenesis. METHODS: A forward genetic screen was performed using Sleeping Beauty transposon insertional mutagenesis to accelerate liver tumour formation in a genetic context in which subtly increased MET RTK levels predispose mice to tumorigenesis. Systematic sequencing of tumours identified common transposon insertion sites, thus uncovering putative RTK cooperators for liver cancer. Bioinformatic analyses were applied to transposon outcomes and human HCC datasets. In vitro and in vivo (through xenografts) functional screens were performed to assess the relevance of distinct cooperative modes to the tumorigenic properties conferred by RTKs. RESULTS: We identified 275 genes, most of which are altered in patients with HCC. Unexpectedly, these genes are not restricted to a small set of pathway/cellular processes, but cover a large spectrum of cellular functions, including signalling, metabolism, chromatin remodelling, mRNA degradation, proteasome, ubiquitination, cell cycle regulation, and chromatid segregation. We validated 15 tumour suppressor candidates, as shRNA-mediated targeting confers tumorigenicity to RTK-sensitized cells, but not to cells with basal RTK levels. This demonstrates that the context of enhanced RTK levels is essential for their action in tumour initiation. CONCLUSION: Our study identifies unanticipated genetic interactions underlying gene cooperativity with RTKs in HCC. Moreover, these results show how subtly increased levels of wild-type RTKs provide a tumour permissive cellular environment allowing a large spectrum of deregulated mechanisms to initiate liver cancer. LAY SUMMARY: Receptor tyrosine kinases (RTKs) are among signals frequently deregulated in patients with hepatocellular carcinoma and their deregulation confers essential biological properties to cancer cells. We have applied a genetic method to randomly mutate large numbers of genes in the context of a mouse model with increased RTK levels, predisposed to develop liver cancer. We identified mechanisms that accelerate tumour formation in cooperation with enhanced RTK levels. The wide array of cellular functions among these cooperators illustrates an extraordinary capability of RTKs to render the liver more vulnerable to additional alterations, by priming cells for tumour initiation.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Hepatocellular , Liver Neoplasms , Liver/pathology , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Mutagenesis, Insertional , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction
5.
Nat Commun ; 9(1): 3976, 2018 09 25.
Article in English | MEDLINE | ID: mdl-30254310

ABSTRACT

In the original version of this Article, the sixth sentence of the abstract incorrectly read 'Most of the genes upregulated and with hypermethylated CGIs in the Alb-R26Met HCC model undergo the same change.', and should have read 'Most of the genes upregulated and with hypermethylated CGIs in the Alb-R26Met HCC model undergo the same change in a large proportion of HCC patients.'. This has been corrected in both the PDF and HTML versions of the Article.

6.
Nat Commun ; 9(1): 3164, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30089774

ABSTRACT

Epigenetic modifications such as aberrant DNA methylation reshape the gene expression repertoire in cancer. Here, we used a clinically relevant hepatocellular carcinoma (HCC) mouse model (Alb-R26Met) to explore the impact of DNA methylation on transcriptional switches associated with tumorigenesis. We identified a striking enrichment in genes simultaneously hypermethylated in CpG islands (CGIs) and overexpressed. These hypermethylated CGIs are located either in the 5'-UTR or in the gene body region. Remarkably, such CGI hypermethylation accompanied by gene upregulation also occurs in 56% of HCC patients, which belong to the "HCC proliferative-progenitor" subclass. Most of the genes upregulated and with hypermethylated CGIs in the Alb-R26Met HCC model undergo the same change. Among reprogrammed genes, several are well-known oncogenes. For others not previously linked to cancer, we demonstrate here their action together as an "oncogene module". Thus, hypermethylation of gene body CGIs is predictive of elevated oncogene levels in cancer, offering a novel stratification strategy and perspectives to normalise cancer gene dosages.


Subject(s)
CpG Islands/genetics , DNA Methylation , Gene Dosage/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Oncogenes/genetics , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Disease Models, Animal , Epigenesis, Genetic , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Up-Regulation
7.
Hepatology ; 66(5): 1644-1661, 2017 11.
Article in English | MEDLINE | ID: mdl-28586114

ABSTRACT

Genetic mutations leading to oncogenic variants of receptor tyrosine kinases (RTKs) are frequent events during tumorigenesis; however, the cellular vulnerability to nononcogenic RTK fluctuations has not been characterized. Here, we demonstrated genetically that in the liver subtle increases in wild-type Met RTK levels are sufficient for spontaneous tumors in mice (Alb-R26Met ), conceptually illustrating how the shift from physiological to pathological conditions results from slight perturbations in signaling dosage. By analyzing 96 different genes in a panel of tumor samples, we demonstrated that liver tumorigenesis modeled by Alb-R26Met mice corresponds to a subset of hepatocellular carcinoma (HCC) patients, thus establishing the clinical relevance of this HCC mouse model. We elucidated the regulatory networks underlying tumorigenesis by combining a phosphokinome screen with bioinformatics analysis. We then used the signaling diversity results obtained from Alb-R26Met HCC versus control livers to design an "educated guess" drug screen, which led to the identification of new, deleterious synthetic lethal interactions. In particular, we report synergistic effects of mitogen-activated protein kinase kinase, ribosomal S6 kinase, and cyclin-dependent kinase 1/2 in combination with Bcl-XL inhibition on a panel of liver cancer cells. Focusing on mitogen-activated protein kinase kinase and Bcl-XL targeting, we mechanistically demonstrated concomitant down-regulation of phosphorylated extracellular signal-regulated kinase and myeloid cell leukemia 1 levels. Of note, a phosphorylated extracellular signal-regulated kinase+/BCL-XL+ /myeloid cell leukemia 1+ signature, deregulated in Alb-R26Met tumors, characterizes a subgroup of HCC patients with poor prognosis. CONCLUSION: Our genetic studies highlight the heightened vulnerability of liver cells to subtle changes in nononcogenic RTK levels, allowing them to acquire a molecular profile that facilitates the full tumorigenic program; furthermore, our outcomes uncover new synthetic lethal interactions as potential therapies for a cluster of HCC patients. (Hepatology 2017;66:1644-1661).


Subject(s)
Carcinoma, Hepatocellular/enzymology , Liver Neoplasms, Experimental/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Carcinogenesis , Carcinoma, Hepatocellular/genetics , Cell Line , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver/enzymology , Liver Neoplasms, Experimental/genetics , Mice, Transgenic , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Molecular Targeted Therapy , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Receptor Protein-Tyrosine Kinases/genetics , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/metabolism
8.
PLoS Genet ; 11(9): e1005533, 2015.
Article in English | MEDLINE | ID: mdl-26393505

ABSTRACT

The successive events that cells experience throughout development shape their intrinsic capacity to respond and integrate RTK inputs. Cellular responses to RTKs rely on different mechanisms of regulation that establish proper levels of RTK activation, define duration of RTK action, and exert quantitative/qualitative signalling outcomes. The extent to which cells are competent to deal with fluctuations in RTK signalling is incompletely understood. Here, we employ a genetic system to enhance RTK signalling in a tissue-specific manner. The chosen RTK is the hepatocyte growth factor (HGF) receptor Met, an appropriate model due to its pleiotropic requirement in distinct developmental events. Ubiquitously enhanced Met in Cre/loxP-based Rosa26(stopMet) knock-in context (Del-R26(Met)) reveals that most tissues are capable of buffering enhanced Met-RTK signalling thus avoiding perturbation of developmental programs. Nevertheless, this ubiquitous increase of Met does compromise selected programs such as myoblast migration. Using cell-type specific Cre drivers, we genetically showed that altered myoblast migration results from ectopic Met expression in limb mesenchyme rather than in migrating myoblasts themselves. qRT-PCR analyses show that ectopic Met in limbs causes molecular changes such as downregulation in the expression levels of Notum and Syndecan4, two known regulators of morphogen gradients. Molecular and functional studies revealed that ectopic Met expression in limb mesenchyme does not alter HGF expression patterns and levels, but impairs HGF bioavailability. Together, our findings show that myoblasts, in which Met is endogenously expressed, are capable of buffering increased RTK levels, and identify mesenchymal cells as a cell type vulnerable to ectopic Met-RTK signalling. These results illustrate that embryonic cells are sensitive to alterations in the spatial distribution of RTK action, yet resilient to fluctuations in signalling levels of an RTK when occurring in its endogenous domain of activity.


Subject(s)
Embryonic Development/genetics , Hepatocyte Growth Factor/genetics , Myoblasts/metabolism , Proto-Oncogene Proteins c-met/genetics , Animals , Cell Movement/genetics , Embryo, Mammalian , Gene Expression Regulation, Developmental , Hepatocyte Growth Factor/metabolism , Mice , Phosphorylation , Proto-Oncogene Proteins c-met/biosynthesis , Signal Transduction , Syndecan-4/biosynthesis , Syndecan-4/genetics
9.
PLoS One ; 7(10): e46738, 2012.
Article in English | MEDLINE | ID: mdl-23071625

ABSTRACT

The development of targeted molecular therapies has provided remarkable advances into the treatment of human cancers. However, in most tumors the selective pressure triggered by anticancer agents encourages cancer cells to acquire resistance mechanisms. The generation of new rationally designed targeting agents acting on the oncogenic path(s) at multiple levels is a promising approach for molecular therapies. 2-phenylimidazo[2,1-b]benzothiazole derivatives have been highlighted for their properties of targeting oncogenic Met receptor tyrosine kinase (RTK) signaling. In this study, we evaluated the mechanism of action of one of the most active imidazo[2,1-b]benzothiazol-2-ylphenyl moiety-based agents, Triflorcas, on a panel of cancer cells with distinct features. We show that Triflorcas impairs in vitro and in vivo tumorigenesis of cancer cells carrying Met mutations. Moreover, Triflorcas hampers survival and anchorage-independent growth of cancer cells characterized by "RTK swapping" by interfering with PDGFRß phosphorylation. A restrained effect of Triflorcas on metabolic genes correlates with the absence of major side effects in vivo. Mechanistically, in addition to targeting Met, Triflorcas alters phosphorylation levels of the PI3K-Akt pathway, mediating oncogenic dependency to Met, in addition to Retinoblastoma and nucleophosmin/B23, resulting in altered cell cycle progression and mitotic failure. Our findings show how the unusual binding plasticity of the Met active site towards structurally different inhibitors can be exploited to generate drugs able to target Met oncogenic dependency at distinct levels. Moreover, the disease-oriented NCI Anticancer Drug Screen revealed that Triflorcas elicits a unique profile of growth inhibitory-responses on cancer cell lines, indicating a novel mechanism of drug action. The anti-tumor activity elicited by 2-phenylimidazo[2,1-b]benzothiazole derivatives through combined inhibition of distinct effectors in cancer cells reveal them to be promising anticancer agents for further investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Molecular Targeted Therapy , Transcriptome/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Benzothiazoles/administration & dosage , Benzothiazoles/adverse effects , Cell Cycle/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Neoplastic/drug effects , Humans , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Mice , Mice, Nude , Mutation, Missense , Phosphorylation , Protein Interaction Maps , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
10.
J Hepatol ; 57(6): 1292-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22889954

ABSTRACT

BACKGROUND & AIMS: Genetic studies indicate that distinct signaling modulators are each necessary but not individually sufficient for embryonic hepatocyte survival in vivo. Nevertheless, how signaling players are interconnected into functional circuits and how they coordinate the balance of cell survival and death in developing livers are still major unresolved issues. In the present study, we examined the modulation of the p53 pathway by HGF/Met in embryonic livers. METHODS: We combined pharmacological and genetic approaches to biochemically and functionally evaluate p53 pathway modulation in primary embryonic hepatocytes and in developing livers. RT-PCR arrays were applied to investigate the selectivity of p53 transcriptional response triggered by Met. RESULTS: Met recruits p53 to regulate the liver developmental program, by qualitatively modulating its transcriptional properties: turning on the Mdm2 survival gene, while keeping death and cell-cycle arrest genes Pmaip1 and p21 silent. We investigated the mechanism leading to p53 regulation by Met and found that Abl and p38MAPK are required for p53 phosphorylation on S(389), Mdm2 upregulation, and hepatocyte survival. Alteration of this signaling mechanism switches p53 properties, leading to p53-dependent cell death in embryonic livers. RT-PCR array studies affirmed the ability of the Met-Abl-p53 axis to modulate the expression of distinct genes that can be regulated by p53. CONCLUSIONS: A signaling circuit involving Abl and p38MAPK is required downstream of Met for the survival of embryonic hepatocytes, via qualitative regulation of the p53 transcriptional response, by switching its proapoptotic into survival properties.


Subject(s)
Hepatocytes/physiology , Liver/embryology , Proto-Oncogene Proteins c-abl/physiology , Proto-Oncogene Proteins c-met/physiology , Transcription, Genetic , Tumor Suppressor Protein p53/physiology , Animals , Cell Survival , Cyclin-Dependent Kinase Inhibitor p21/genetics , Mice , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-mdm2/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
J Neurosci ; 31(31): 11144-58, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21813676

ABSTRACT

The precise control of motor neuron (MN) death and survival following initial innervation of skeletal muscle targets is a key step in sculpting a functional motor system, but how this is regulated at the level of individual motor pools remains unclear. Hepatocyte growth factor (HGF) and its receptor Met play key developmental roles in both muscle and MNs. We generated mice (termed "Nes-Met") in which met is inactivated from midembryonic stages onward in the CNS only. Adult animals showed motor behavioral defects suggestive of impaired innervation of pectoral muscles. Correspondingly, in neonatal spinal cords of Nes-Met mutants, we observed death of a discrete population of pea3-expressing MNs at brachial levels. Axonal tracing using pea3 reporter mice revealed a novel target muscle of pea3-expressing MNs: the pectoralis minor muscle. In Nes-Met mice, the pectoralis minor pool initially innervated its target muscle, but required HGF/Met for survival, hence for proper maintenance of muscle innervation. In contrast, HGF/Met was dispensable for the survival of neighboring Met-expressing MN pools, despite its earlier functions for their specification and axon growth. Our results demonstrate the exquisite degree to which outcomes of signaling by receptor tyrosine kinases are regulated on a cell-by-cell basis. They also provide a model for one way in which the multiplicity of neurotrophic factors may allow for regulation of MN numbers in a pool-specific manner.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Motor Neurons/drug effects , Motor Neurons/physiology , Nerve Growth Factors/pharmacology , Animals , Animals, Newborn , Axons/physiology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Choline O-Acetyltransferase/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Dose-Response Relationship, Drug , Embryo, Mammalian , Ganglia, Spinal/cytology , Gene Expression Regulation, Developmental/genetics , Green Fluorescent Proteins/genetics , Hand Strength/physiology , Hepatocyte Growth Factor/genetics , Intermediate Filament Proteins/genetics , Male , Mice , Mice, Transgenic , Models, Biological , Motor Activity/genetics , Muscle Strength/genetics , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Mutation/genetics , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Nestin , Proto-Oncogene Proteins c-ets/genetics , Proto-Oncogene Proteins c-met/genetics , Psychomotor Performance/physiology , Reaction Time , Rotarod Performance Test , Spinal Cord/cytology , Transcription Factors/genetics
12.
Immunity ; 31(2): 197-208, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19682930

ABSTRACT

Despite compromised T cell antigen receptor (TCR) signaling, mice in which tyrosine 136 of the adaptor linker for activation of T cells (LAT) was constitutively mutated (Lat(Y136F) mice) accumulate CD4(+) T cells that trigger autoimmunity and inflammation. Here we show that equipping postthymic CD4(+) T cells with LATY136F molecules or rendering them deficient in LAT molecules triggers a lymphoproliferative disorder dependent on prior TCR engagement. Therefore, such disorders required neither faulty thymic T cell maturation nor LATY136F molecules. Unexpectedly, in CD4(+) T cells recently deprived of LAT, the proximal triggering module of the TCR induced a spectrum of protein tyrosine phosphorylation that largely overlapped the one observed in the presence of LAT. The fact that such LAT-independent signals result in lymphoproliferative disorders with excessive cytokine production demonstrates that LAT constitutes a key negative regulator of the triggering module and of the LAT-independent branches of the TCR signaling cassette.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Lymphoproliferative Disorders/immunology , Membrane Proteins/immunology , Phosphoproteins/immunology , Receptors, Antigen, T-Cell/immunology , Adaptor Proteins, Signal Transducing/genetics , Adoptive Transfer , Animals , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cytokines/immunology , Cytokines/metabolism , Histocompatibility Antigens Class II/metabolism , Lymphoproliferative Disorders/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mutation , Phosphoproteins/genetics , Phosphorylation/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/immunology
13.
J Immunol ; 180(3): 1565-75, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18209052

ABSTRACT

Mutant mice where tyrosine 136 of linker for activation of T cells (LAT) was replaced with a phenylalanine (Lat(Y136F) mice) develop a fast-onset lymphoproliferative disorder involving polyclonal CD4 T cells that produce massive amounts of Th2 cytokines and trigger severe inflammation and autoantibodies. We analyzed whether the Lat(Y136F) pathology constitutes a bona fide autoimmune disorder dependent on TCR specificity. Using adoptive transfer experiments, we demonstrated that the expansion and uncontrolled Th2-effector function of Lat(Y136F) CD4 cells are not triggered by an MHC class II-driven, autoreactive process. Using Foxp3EGFP reporter mice, we further showed that nonfunctional Foxp3(+) regulatory T cells are present in Lat(Y136F) mice and that pathogenic Lat(Y136F) CD4 T cells were capable of escaping the control of infused wild-type Foxp3(+) regulatory T cells. These results argue against a scenario where the Lat(Y136F) pathology is primarily due to a lack of functional Foxp3(+) regulatory T cells and suggest that a defect intrinsic to Lat(Y136F) CD4 T cells leads to a state of TCR-independent hyperactivity. This abnormal status confers Lat(Y136F) CD4 T cells with the ability to trigger the production of Abs and of autoantibodies in a TCR-independent, quasi-mitogenic fashion. Therefore, despite the presence of autoantibodies causative of severe systemic disease, the pathological conditions observed in Lat(Y136F) mice unfold in an Ag-independent manner and thus do not qualify as a genuine autoimmune disorder.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autoimmune Diseases/immunology , Lymphoproliferative Disorders/immunology , Membrane Proteins/genetics , Phosphoproteins/genetics , T-Lymphocytes, Regulatory/immunology , Th2 Cells/immunology , Animals , Autoantibodies/blood , Autoimmune Diseases/genetics , CD4 Antigens/analysis , Cell Proliferation , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/genetics , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Histocompatibility Antigens Class II/immunology , Interleukin-7/metabolism , Lymphoproliferative Disorders/genetics , Mice , Mice, Mutant Strains , Receptors, Antigen, T-Cell/immunology
14.
Eur J Immunol ; 37(11): 3259-69, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17918199

ABSTRACT

LIME (Lck-interacting membrane protein) is a transmembrane adaptor that associates with the Lck and Fyn protein tyrosine kinases and with the C-terminal Src kinase (Csk). To delineate the role of LIME in vivo, LIME-deficient mice were generated. Although Lime transcripts were expressed in immature and mature B and T cells, the absence of LIME impeded neither the development nor the function of B and T cells. TCR transgenic mice deprived of LIME showed, however, a 1.8-fold enhancement in positive selection. Since B cells and activated T cells express LIME and the related adaptor NTAL, mice lacking both adaptors were generated. Double-deficient mice showed no defect in the development and function of B and T cells, and the lack of LIME had no effect on the autoimmune syndrome that develops in aged NTAL-deficient mice. In contrast to a previous report, we further showed that this autoimmune syndrome develops in the absence of T cells. Therefore, our in vivo results refute all the previous roles postulated for LIME on the basis of studies of transformed B and T cells and demonstrate that LIME has no seminal role in the signaling cassette operated by antigen receptors and coreceptors.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , B-Lymphocytes/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Vesicular Transport/deficiency , Animals , Autoimmunity , B-Lymphocytes/cytology , Blotting, Western , Cell Differentiation/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/cytology
15.
Nat Immunol ; 4(10): 999-1008, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12970761

ABSTRACT

LAT (linker for activation of T cells) is essential for T cell receptor signaling. Mice homozygous for a mutation of the three C-terminal LAT tyrosine residues showed a block in alphabeta T cell development and a partially impaired gammadelta T cell development. Without intentional immunization, they accumulated gammadelta T cells in the spleen and lymph nodes that chronically produced T helper type 2 cytokines in large amounts, and caused the maturation of plasma cells secreting immunoglobulin E (IgE) and IgG1. These effects are very similar to that triggered in the alphabeta lineage by a mutation involving a distinct LAT tyrosine. Thus, LAT is an essential regulator of T cell homeostasis and terminal differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/immunology , Membrane Proteins , Phosphoproteins/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Base Sequence , Carrier Proteins/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cytokines/immunology , Flow Cytometry , Homeostasis/immunology , Immunohistochemistry , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Sequence Data , Mutation , Phenotype , Phosphoproteins/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Spleen/immunology , Th2 Cells/immunology , Th2 Cells/metabolism , Tyrosine/genetics
16.
Science ; 296(5575): 2036-40, 2002 Jun 14.
Article in English | MEDLINE | ID: mdl-12065839

ABSTRACT

The transmembrane protein LAT (linker for activation of T cells) couples the T cell receptor (TCR) to downstream signaling effectors. Mice homozygous for a mutation of a single LAT tyrosine residue showed impeded T cell development. However, later they accumulated polyclonal helper T (TH) cells that chronically produced type 2 cytokines in large amounts. This exaggerated TH2 differentiation caused tissue eosinophilia and massive maturation of plasma cells secreting to immunoglobulins of the E and G1 isotypes. This paradoxical phenotype establishes an unanticipated inhibitory function for LAT that is critical for the differentiation and homeostasis of TH cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/genetics , Carrier Proteins/physiology , Membrane Proteins , Phosphoproteins/genetics , Phosphoproteins/physiology , Point Mutation , Th2 Cells/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/physiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/physiology , CD5 Antigens/analysis , CD5 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Cycle , Cell Differentiation , Eosinophilia , Eosinophils/physiology , Histocompatibility Antigens Class II/immunology , Immunoglobulin E/blood , Immunoglobulin G/blood , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukins/genetics , Interleukins/metabolism , Leukocyte Count , Lymphocyte Activation , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Mice, Inbred BALB C , Phenotype , Receptors, Antigen, T-Cell/analysis , Signal Transduction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/physiology , Th2 Cells/physiology , Thymus Gland/cytology , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...