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










Publication year range
1.
iScience ; 23(3): 100959, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32179481

ABSTRACT

Defining the pathways that control cardiac development facilitates understanding the pathogenesis of congenital heart disease. Herein, we identify enrichment of a Cullin5 Ub ligase key subunit, Asb2, in myocardial progenitors and differentiated cardiomyocytes. Using two conditional murine knockouts, Nkx+/Cre.Asb2fl/fl and AHF-Cre.Asb2fl/fl, and tissue clarifying technique, we reveal Asb2 requirement for embryonic survival and complete heart looping. Deletion of Asb2 results in upregulation of its target Filamin A (Flna), and concurrent Flna deletion partially rescues embryonic lethality. Conditional AHF-Cre.Asb2 knockouts harboring one Flna allele have double outlet right ventricle (DORV), which is rescued by biallelic Flna excision. Transcriptomic and immunofluorescence analyses identify Tgfß/Smad as downstream targets of Asb2/Flna. Finally, using CRISPR/Cas9 genome editing, we demonstrate Asb2 requirement for human cardiomyocyte differentiation suggesting a conserved mechanism between mice and humans. Collectively, our study provides deeper mechanistic understanding of the role of the ubiquitin proteasome system in cardiac development and suggests a previously unidentified murine model for DORV.

2.
Trends Mol Med ; 25(9): 760-774, 2019 09.
Article in English | MEDLINE | ID: mdl-31235369

ABSTRACT

For long-lived contractile cells, such as striated muscle cells, maintaining proteome integrity is a challenging task. These cells require hundreds of components that must be properly synthesized, folded, and incorporated into the basic contractile unit, the sarcomere. Muscle protein quality control in cells is mainly guaranteed by the ubiquitin-proteasome system (UPS), the lysosome-autophagy system, and various molecular chaperones. Recent studies establish the concept of dedicated UPS in the regulation of sarcomere assembly during development and in adult life to maintain the intricate and interwoven organization of protein complexes in muscle. Failure of sarcomere protein quality control often represents the basis of severe myopathies and cardiomyopathies in human, further highlighting its importance in producing and maintaining the contractile machinery of muscle cells in shape.


Subject(s)
Disease Susceptibility , Homeostasis , Muscle, Striated/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Autophagy , Humans , Molecular Chaperones/metabolism , Muscle, Striated/pathology , Protein Binding , Sarcomeres , Signal Transduction , Ubiquitination
3.
Cancer Immunol Res ; 7(8): 1332-1344, 2019 08.
Article in English | MEDLINE | ID: mdl-31175139

ABSTRACT

The escape of cancer cells from host immunosurveillance involves a shift in immune responses, including an imbalance in Th1 and Th2 cells. A Th1-dominated immune response predicts positive outcomes in colorectal cancer. The E3 ubiquitin ligase, Asb2α, is expressed in Th2 cells, but its roles in T-cell maturation and cancer are unclear. We provide evidence that the Th2 master regulator, Gata3, induces Asb2 Loss of Asb2 did not affect Th differentiation ex vivo, but reduced IL4 production from Th2 cells. We found that high ASB2 expression was associated with poor outcome in colorectal cancer. Loss of Asb2 from hematopoietic cells promoted a Th1 response and attenuated colitis-associated tumorigenesis in mice. Diminished Th2 function correlated with increased IFNγ production and an enhanced type 1 antitumor immune response in Asb2-deficient mice. Our work suggests that Asb2α promotes a Th2 phenotype in vivo, which in turn is associated with tumor progression in a mouse model of colitis.


Subject(s)
Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Immunomodulation , Th2 Cells/immunology , Th2 Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Binding Sites , Cell Differentiation/genetics , Cell Differentiation/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Computational Biology/methods , Databases, Genetic , Gene Expression Profiling , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Mice , Protein Binding , Recurrence , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
4.
Circ Res ; 122(6): e34-e48, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29374072

ABSTRACT

RATIONALE: Heart development involves differentiation of cardiac progenitors and assembly of the contractile sarcomere apparatus of cardiomyocytes. However, little is known about the mechanisms that regulate actin cytoskeleton remodeling during cardiac cell differentiation. OBJECTIVE: The Asb2α (Ankyrin repeat-containing protein with a suppressor of cytokine signaling box 2) CRL5 (cullin 5 RING E3 ubiquitin ligase) triggers polyubiquitylation and subsequent degradation by the proteasome of FLNs (filamins). Here, we investigate the role of Asb2α in heart development and its mechanisms of action. METHODS AND RESULTS: Using Asb2 knockout embryos, we show that Asb2 is an essential gene, critical to heart morphogenesis and function, although its loss does not interfere with the overall patterning of the embryonic heart tube. We show that the Asb2α E3 ubiquitin ligase controls Flna stability in immature cardiomyocytes. Importantly, Asb2α-mediated degradation of the actin-binding protein Flna marks a previously unrecognized intermediate step in cardiac cell differentiation characterized by cell shape changes and actin cytoskeleton remodeling. We further establish that in the absence of Asb2α, myofibrils are disorganized and that heartbeats are inefficient, leading to embryonic lethality in mice. CONCLUSIONS: These findings identify Asb2α as an unsuspected key regulator of cardiac cell differentiation and shed light on the molecular and cellular mechanisms determining the onset of myocardial cell architecture and its link with early cardiac function. Although Flna is known to play roles in cytoskeleton organization and to be required for heart function, this study now reveals that its degradation mediated by Asb2α ensures essential functions in differentiating cardiac progenitors.


Subject(s)
Actin Cytoskeleton/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Filamins/metabolism , Heart/growth & development , Myocytes, Cardiac/metabolism , Ubiquitination , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Differentiation , Cells, Cultured , Filamins/genetics , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/cytology , Proteolysis , Suppressor of Cytokine Signaling Proteins
5.
Biochimie ; 122: 339-47, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26253693

ABSTRACT

Ubiquitylation is a reversible post-translational modification of proteins that controls a myriad of functions and cellular processes. It occurs through the sequential action of three distinct enzymes. E3 ubiquitin ligases (E3s) play the role of conductors of the ubiquitylation pathway making them attractive therapeutic targets. This review is dedicated to the largest family of multimeric E3s, the Cullin-RING E3 (CRL) family and more specifically to cullin 5 based CRLs that remains poorly characterized.


Subject(s)
Cullin Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Antineoplastic Agents/therapeutic use , Humans , Models, Biological , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction/drug effects , Ubiquitin/metabolism
6.
Sci Rep ; 5: 16269, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26537633

ABSTRACT

Conventional dendritic cells (cDCs) comprise distinct populations with specialized immune functions that are mediators of innate and adaptive immune responses. Transcriptomic and proteomic approaches have been used so far to identify transcripts and proteins that are differentially expressed in these subsets to understand the respective functions of cDCs subsets. Here, we showed that the Cullin 5-RING E3 ubiquitin ligase (E3) ASB2α, by driving degradation of filamin A (FLNa) and filamin B (FLNb), is responsible for the difference in FLNa and FLNb abundance in the different spleen cDC subsets. Importantly, the ability of these cDC subsets to migrate correlates with the level of FLNa. Furthermore, our results strongly point to CD4 positive and double negative cDCs as distinct populations. Finally, we develop quantitative global proteomic approaches to identify ASB2α substrates in DCs using ASB2 conditional knockout mice. As component of the ubiquitin-proteasome system (UPS) are amenable to pharmacological manipulation, these approaches aimed to the identification of E3 substrates in physiological relevant settings could potentially lead to novel targets for therapeutic strategies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dendritic Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line, Tumor , Filamins/metabolism , HeLa Cells , Humans , Mice , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism , Proteomics/methods , Suppressor of Cytokine Signaling Proteins , Ubiquitin/metabolism
7.
J Mol Cell Cardiol ; 87: 214-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26343497

ABSTRACT

BACKGROUND: Hypertrophic cardiomyopathy (HCM) is an autosomal-dominant disease with mutations in genes encoding sarcomeric proteins. Previous findings suggest deregulation of the ubiquitin proteasome system (UPS) in HCM in humans and in a mouse model of HCM (Mybpc3-targeted knock-in (KI) mice). In this study we investigated transcript levels of several muscle-specific E3 ubiquitin ligases in KI mice and aimed at identifying novel protein targets. METHODS AND RESULTS: Out of 9 muscle-specific E3 ligases, Asb2ß was found with the lowest mRNA level in KI compared to wild-type (WT) mice. After adenoviral-mediated Asb2ß transduction of WT neonatal mouse cardiomyocytes with either a WT or inactive Asb2ß mutant, desmin was identified as a new target of Asb2ß by mass spectrometry, co-immunoprecipitation and immunoblotting. Immunofluorescence analysis revealed a co-localization of desmin with Asb2ß at the Z-disk of the sarcomere. Knock-down of Asb2ß in cardiomyocytes resulted in higher desmin protein levels. Furthermore, desmin levels were higher in ventricular samples of HCM mice and patients than controls. CONCLUSIONS: This study identifies desmin as a new Asb2ß target for proteasomal degradation in cardiomyocytes and suggests that accumulation of desmin could contribute to UPS impairment in HCM mice and patients.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cardiomyopathy, Hypertrophic/genetics , Desmin/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Cardiomyopathy, Hypertrophic/pathology , Desmin/genetics , Disease Models, Animal , Gene Expression Regulation , Humans , Mice , Mutation , Myocardium/pathology , Myocytes, Cardiac/pathology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Sarcomeres/metabolism , Suppressor of Cytokine Signaling Proteins , Ubiquitin
8.
Cell Signal ; 25(12): 2823-30, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24044920

ABSTRACT

ASB proteins are the specificity subunits of cullin5-RING E3 ubiquitin ligases (CRL5) that play roles in ubiquitin-mediated protein degradation. However, how their activity is regulated remains poorly understood. Here, we unravel a novel mechanism of regulation of a CRL5 through phosphorylation of its specificity subunit ASB2α. Indeed, using mass spectrometry, we showed for the first time that ASB2α is phosphorylated and that phosphorylation of serine-323 (Ser-323) of ASB2α is crucial for the targeting of the actin-binding protein filamin A (FLNa) to degradation. Mutation of ASB2α Ser-323 to Ala had no effect on intrinsic E3 ubiquitin ligase activity of ASB2α but abolished the ability of ASB2α to induce degradation of FLNa. In contrast, the ASB2α Ser-323 to Asp phosphomimetic mutant induced acute degradation of FLNa. Moreover, inhibition of the extracellular signal-regulated kinases 1 and 2 (Erk1/2) activity reduced ASB2α-mediated FLNa degradation. We further showed that the subcellular localization of ASB2α to actin-rich structures is dependent on ASB2α Ser-323 phosphorylation and propose that the interaction with FLNa depends on the electrostatic potential redistribution induced by the Ser-323 phosphate group. Taken together, these data unravel an important mechanism by which ASB2α-mediated FLNa degradation can be regulated.


Subject(s)
Filamins/metabolism , Proteolysis , Serine/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Amino Acid Sequence , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Models, Molecular , Molecular Sequence Data , Phosphorylation , Serine/analysis , Suppressor of Cytokine Signaling Proteins/chemistry , Ubiquitin-Protein Ligases/metabolism
9.
Blood ; 122(4): 533-41, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23632887

ABSTRACT

The actin-binding protein filamins (FLNs) are major organizers of the actin cytoskeleton. They control the elasticity and stiffness of the actin network and provide connections with the extracellular microenvironment by anchoring transmembrane receptors to the actin filaments. Although numerous studies have revealed the importance of FLN levels, relatively little is known about the regulation of its stability in physiological relevant settings. Here, we show that the ASB2α cullin 5-ring E3 ubiquitin ligase is highly expressed in immature dendritic cells (DCs) and is down-regulated after DC maturation. We further demonstrate that FLNs are substrates of ASB2α in immature DCs and therefore are not stably expressed in these cells, whereas they exhibit high levels of expression in mature DCs. Using ASB2 conditional knockout mice, we show that ASB2α is a critical regulator of cell spreading and podosome rosette formation in immature DCs. Furthermore, we show that ASB2(-/-) immature DCs exhibit reduced matrix-degrading function leading to defective migration. Altogether, our results point to ASB2α and FLNs as newcomers in DC biology.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Movement/genetics , Dendritic Cells/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Contractile Proteins/genetics , Contractile Proteins/metabolism , Dendritic Cells/metabolism , Filamins , Gene Knockdown Techniques , Granulocyte-Macrophage Progenitor Cells/metabolism , Granulocyte-Macrophage Progenitor Cells/physiology , Isoenzymes/genetics , Isoenzymes/metabolism , Isoenzymes/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , NIH 3T3 Cells , Suppressor of Cytokine Signaling Proteins , Transfection , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/physiology
10.
J Biol Chem ; 286(35): 30571-30581, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21737450

ABSTRACT

By providing contacts between hematopoietic cells and the bone marrow microenvironment, integrins are implicated in cell adhesion and thereby in control of cell fate of normal and leukemia cells. The ASB2 gene, initially identified as a retinoic acid responsive gene and a target of the promyelocytic leukemia retinoic acid receptor α oncoprotein in acute promyelocytic leukemia cells, encodes two isoforms, a hematopoietic-type (ASB2α) and a muscle-type (ASB2ß) that are involved in hematopoietic and myogenic differentiation, respectively. ASB2α is the specificity subunit of an E3 ubiquitin ligase complex that targets filamins to proteasomal degradation. To examine the relationship of the ASB2α structure to E3 ubiquitin ligase function, functional assays and molecular modeling were performed. We show that ASB2α, through filamin A degradation, enhances adhesion of hematopoietic cells to fibronectin, the main ligand of ß1 integrins. Furthermore, we demonstrate that a short N-terminal region specific to ASB2α, together with ankyrin repeats 1 to 10, is necessary for association of ASB2α with filamin A. Importantly, the ASB2α N-terminal region comprises a 9-residue segment with predicted structural homology to the filamin-binding motifs of migfilin and ß integrins. Together, these data provide new insights into the molecular mechanisms of ASB2α binding to filamin.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Integrins/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Animals , Cell Adhesion , Fibronectins/metabolism , HeLa Cells , Humans , Mice , Muscles/metabolism , NIH 3T3 Cells , Protein Binding , Protein Structure, Tertiary , Substrate Specificity
11.
Blood ; 112(13): 5130-40, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18799729

ABSTRACT

The ordered series of proliferation and differentiation from hematopoietic progenitor cells is disrupted in leukemia, resulting in arrest of differentiation at immature proliferative stages. Characterizing the molecular basis of hematopoietic differentiation is therefore important for understanding and treating disease. Retinoic acid induces expression of ankyrin repeat-containing protein with a suppressor of cytokine signaling box 2 (ASB2) in acute promyelocytic leukemia cells, and ASB2 expression inhibits growth and promotes commitment, recapitulating an early step critical for differentiation. ASB2 is the specificity subunit of an E3 ubiquitin ligase complex and is proposed to exert its effects by regulating the turnover of specific proteins; however, no ASB2 substrates had been identified. Here, we report that ASB2 targets the actin-binding proteins filamin A and B for proteasomal degradation. Knockdown of endogenous ASB2 in leukemia cells delays retinoic acid-induced differentiation and filamin degradation; conversely, ASB2 expression in leukemia cells induces filamin degradation. ASB2 expression inhibits cell spreading, and this effect is recapitulated by knocking down both filamin A and filamin B. Thus, we suggest that ASB2 may regulate hematopoietic cell differentiation by modulating cell spreading and actin remodeling through targeting of filamins for degradation.


Subject(s)
Contractile Proteins/metabolism , Leukemia/pathology , Microfilament Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Suppressor of Cytokine Signaling Proteins/physiology , Actins/metabolism , Cell Adhesion , Cell Differentiation/drug effects , Cell Line, Tumor , Contractile Proteins/genetics , Filamins , Humans , Leukemia/drug therapy , Microfilament Proteins/genetics , RNA, Small Interfering/pharmacology , Suppressor of Cytokine Signaling Proteins/genetics , Tretinoin/pharmacology
12.
Blood Cells Mol Dis ; 40(2): 200-10, 2008.
Article in English | MEDLINE | ID: mdl-17919948

ABSTRACT

Understanding the molecular mechanisms controlling normal hematopoietic differentiation is critical to develop new treatments for blood diseases and to manipulate stem cells. Despite the identification of many players in hematopoiesis, the molecular mechanisms controlling hematopoietic differentiation remain poorly understood. Due to a number of recent findings, the targeting of regulators of hematopoiesis to proteasomal degradation might be an important step in control of this developmental program.


Subject(s)
Hematologic Neoplasms/metabolism , Hematopoiesis , Hematopoietic System/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing , Humans , Intracellular Signaling Peptides and Proteins , Proteins/metabolism , RING Finger Domains/physiology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases/metabolism
13.
Mol Cell Biol ; 26(16): 6209-22, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16880530

ABSTRACT

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase, initially discovered as part of the NPM-ALK fusion protein, resulting from the t(2;5) translocation that is frequently associated with anaplastic large-cell lymphomas. The native ALK protein is normally expressed in the developing and, at a weaker level, adult nervous system. We recently demonstrated that the oncogenic, constitutively kinase-activated NPM-ALK protein was antiapoptotic when expressed in Jurkat lymphoblastic cells treated with cytotoxic drugs. In contrast, we now show that Jurkat cells overexpressing the wild-type ALK receptor are more sensitive to doxorubicin-induced apoptosis than parental cells. Moreover, the ALK protein is cleaved during apoptosis in a caspase-dependent manner. Mutation of aspartic residues to asparagine allowed us to map the caspase cleavage site in the juxtamembrane region of ALK. In order to assess the role of ALK in neural cell-derived tissue, we transiently expressed ALK in the 13.S.1.24 rat neuroblast immortalized cell line. ALK expression led to apoptotic cell death of the neuroblasts. ALK ligation by specific activating antibodies decreased ALK-facilitated apoptosis in both lymphoid and neuronal cell lines. Moreover, ALK transfection reduced the survival of primary cultures of cortical neurons. Thus, ALK has a proapoptotic activity in the absence of ligand, whereas it is antiapoptotic in the presence of its ligand and when the kinase is intrinsically activated. These properties place ALK in the growing family of dependence receptors.


Subject(s)
Apoptosis , Caspases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/metabolism , Anaplastic Lymphoma Kinase , Animals , Antibodies/immunology , Apoptosis/drug effects , Aspartic Acid/genetics , Caspase 3 , Cell Line, Tumor , Cell Membrane/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/enzymology , Doxorubicin/pharmacology , Enzyme Activation , Gene Expression , Humans , Jurkat Cells , Mice , Mutation/genetics , Neurons/cytology , Neurons/enzymology , Protein Processing, Post-Translational , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases , Transfection
14.
J Cell Sci ; 118(Pt 24): 5811-23, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16317043

ABSTRACT

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase essentially and transiently expressed in specific areas of the developing central and peripheral nervous systems. We previously demonstrated that a membrane-bound and constitutively active form of the ALK protein tyrosine kinase (PTK) domain induced the neuron-like differentiation of PC12 cells through specific activation of the mitogen-activated protein kinase (MAP kinase) pathway. Its PTK domain had been originally identified in a nucleo-cytosolic and constitutively active transforming protein, NPM-ALK. Downstream targets involved in oncogenic proliferation and survival processes have been proposed to include phospholipase Cgamma (PLCgamma), phosphoinositide 3-kinase (PI 3-kinase)/AKT, STAT 3/5 and Src. We therefore postulated that activation of specific signaling pathways leading to differentiation or proliferation can be differently controlled depending on the subcellular localization of ALK PTK domain. To increase knowledge of its physiological role in the nervous system, we focused in the present study on the influence of its subcellular localization on neuronal differentiation. To achieve this goal, we characterized biological responses and transduction pathways in PC12 cells elicited by various constructs encoding membrane-bound (through transmembrane or myristyl sequences) or cytosolic ALK-derived proteins. In order to control the activation of their PTK domain, we used an inducible dimerization system. Here, we demonstrate that membrane attachment of the ALK PTK domain, in PC12 cells, is crucial for initiation of neurite outgrowth and proliferation arrest through a decrease of DNA synthesis. Furthermore, we show that this differentiation process relies on specific and sustained activation of ERK 1/2 proteins. By contrast, activation of the cytosolic form of this domain fails to induce MAP kinase activation and cell differentiation but promotes a PI 3-kinase/AKT-dependent PC12 cell proliferation. These data indicate that subcellular localization of the ALK PTK domain was a determinant for the control and specificity of downstream transduction cascades and was crucial for deciding the fate to which the neuronal cell will be committed.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Neurons/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Anaplastic Lymphoma Kinase , Animals , Dimerization , Neurons/cytology , PC12 Cells , Phosphotransferases/genetics , Phosphotransferases/metabolism , Protein Structure, Tertiary , Protein-Tyrosine Kinases/genetics , Rats , Receptor Protein-Tyrosine Kinases
15.
J Biol Chem ; 280(28): 26039-48, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15886198

ABSTRACT

Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase that is transiently expressed in specific regions of the central and peripheral nervous systems, suggesting a role in its normal development and function. The nature of the cognate ligands of ALK in vertebrate is still a matter of debate. We produced a panel of monoclonal antibodies (mAbs) directed against the extracellular domain of the human receptor. Two major species of ALK (220 and 140 kDa) were identified in transfected cells, and the use of our mAbs established that the 140-kDa species results from a cleavage of the 220-kDa form. Two mAbs, in the nm range, induced the differentiation of PC12 cells transiently transfected with ALK. In human embryonic kidney 293 cells stably expressing ALK, these two mAbs strongly activated the receptor and subsequently the mitogen-activated protein kinase pathway. We further showed for the first time that activation of ALK also resulted in a specific activation of STAT3. In contrast, other mAbs presented the characteristics of blocking antibodies. Finally, in these cell systems, a mitogenic form of pleiotrophin, a proposed ligand of ALK, failed to activate this receptor. Thus, in the absence of clearly established ligand(s) in vertebrates, the availability of mAbs allowing the activation or the inhibition of the receptor will be essential for a better understanding of the biological roles of ALK.


Subject(s)
Antibodies, Monoclonal/chemistry , Carrier Proteins/pharmacology , Cytokines/pharmacology , Enzyme Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase , Animals , Carrier Proteins/chemistry , Cell Line , Cytokines/chemistry , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Ligands , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Models, Biological , Neurons/metabolism , PC12 Cells , Phosphorylation , Plasmids/metabolism , Protein Structure, Tertiary , Rats , Receptor Protein-Tyrosine Kinases , Time Factors , Transfection , Tyrosine/chemistry
16.
Blood ; 102(9): 3371-8, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-12869515

ABSTRACT

Retinoic acid induces clinical remission in acute promyelocytic leukemia (APL) by triggering differentiation of leukemia promyelocytes. Here, we have characterized a gene encoding a member of the immunoglobulin superfamily, among novel retinoic acid-induced genes identified in APL cells. This protein, which was named JAML (junctional adhesion molecule-like), contains 2 extracellular immunoglobulin-like domains, a transmembrane segment, and a cytoplasmic tail. JAML mRNA is expressed in hematopoietic tissues and is prominently expressed in granulocytes. The fact that JAML protein is localized at the cell plasma membrane in the areas of cell-cell contacts, whereas it is not detected at free cell borders, suggests that JAML is engaged in homophilic interactions. Furthermore, a conserved dimerization motif among JAM members was shown to be important for JAML localization at the cell membrane. Finally, exogenous expression of JAML in myeloid leukemia cells resulted in enhanced cell adhesion to endothelial cells. Altogether, our results point to JAML as a novel member of the JAM family expressed on leukocytes with a possible role in leukocyte transmigration.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Gene Expression Regulation/drug effects , Leukemia, Myeloid/pathology , Base Sequence , Cell Adhesion/drug effects , Cell Adhesion Molecules/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Endothelium, Vascular/cytology , Humans , Junctional Adhesion Molecules , Leukemia, Myeloid/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Molecular Sequence Data , RNA, Messenger/biosynthesis , Sequence Analysis , Tretinoin/pharmacology , Tumor Cells, Cultured
17.
Oncogene ; 22(24): 3770-80, 2003 Jun 12.
Article in English | MEDLINE | ID: mdl-12802284

ABSTRACT

MLN64, is invariably coamplified and coexpressed with erbB-2 in breast cancers. The human MLN64 and ERBB2 genes are positioned at less than 50 kb from each other, on chromosome 17q12. To understand the molecular basis of MLN64 overexpression in cancer, the genomic region containing the MLN64 and ERBB2 genes was isolated and mapped. The two genes, DARPP32 and Telethonin, flanking MLN64 respectively on its centromeric and telomeric sides, although coamplified, are not overexpressed in breast cancer cells, indicating that gene amplification is not sufficient to allow overexpression. The MLN64 minimal promoter was isolated and found to be a housekeeping gene promoter containing four potential Sp1 binding elements. Using Sp1-deficient Drosophila SL2 cells, MLN64 promoter activity was induced in a dose-dependent manner by exogenous Sp1 addition. Furthermore, mutation of each individual Sp1 element resulted in a significant decrease in reporter gene activity, indicating that all the Sp1 binding elements are functional and act together to promote gene expression. Since the ERBB2 promoter is also positively regulated by Sp1, this study indicates that MLN64 and ERBB2 genes share common transcriptional controls together with a physical link on chromosome 17q. We speculate that, in addition to the oncogenic potential of erbB-2 overexpression, the unbalanced action of MLN64 contributes to the poor clinical outcome of breast tumors bearing this amplified region.


Subject(s)
Breast Neoplasms/genetics , Carrier Proteins , Gene Expression Regulation, Neoplastic , Membrane Proteins/genetics , Sp1 Transcription Factor/physiology , Amino Acid Sequence , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Protein-beta/physiology , Cell Line , Conserved Sequence , Female , Gene Amplification , Genes, erbB-2 , Humans , Membrane Proteins/chemistry , Molecular Sequence Data , Promoter Regions, Genetic
18.
J Biol Chem ; 277(1): 218-24, 2002 Jan 04.
Article in English | MEDLINE | ID: mdl-11682484

ABSTRACT

In acute promyelocytic leukemia (APL) cells harboring the promyelocytic leukemia retinoic acid receptor alpha (PML-RARalpha) chimeric protein, retinoic acid (RA)-induced differentiation is triggered through a PML-RARalpha signaling resulting in activation of critical target genes. Induced differentiation of APL cells is always preceded by withdrawal from the cell cycle and commitment events leading to terminal differentiation. Here we have identified the human ankyrin repeat-containing protein with a suppressor of cytokine signaling box-2 (ASB-2) cDNA, as a novel RA-induced gene in APL cells. PML-RARalpha strongly enhanced RA-induced ASB-2 mRNA expression. In myeloid leukemia cells, ASB-2 expression induced growth inhibition and chromatin condensation recapitulating early events critical to RA-induced differentiation of APL cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/physiology , Leukemia, Promyelocytic, Acute/pathology , Amino Acid Sequence , Base Sequence , Carrier Proteins/genetics , Cell Differentiation/drug effects , Chromatin/metabolism , Humans , Molecular Sequence Data , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/physiology , RNA, Messenger/analysis , Suppressor of Cytokine Signaling Proteins , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...