Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Biotechnol Bioeng ; 119(1): 287-298, 2022 01.
Article in English | MEDLINE | ID: mdl-34708875

ABSTRACT

As protein-protein interactions (PPIs) are involved in many cellular events, development of mammalian cytosolic PPI detection systems is important for drug discovery as well as understanding biological phenomena. We have previously reported a c-kit-based PPI screening (KIPPIS) system, in which proteins of interest were fused with a receptor tyrosine kinase c-kit, leading to intracellular PPI-dependent cell growth. However, it has not been investigated whether PPI can be detected using other receptors. In this study, we employed a thrombopoietin receptor, which belongs to the Type I cytokine receptor family, to develop a thrombopoietin receptor-based PPI screening (THROPPIS) system. To improve the sensitivity of THROPPIS, we examined two strategies of (i) localization of the chimeric receptors on the cell membrane, and (ii) addition of a helper module to the chimeric receptors. Intriguingly, the nonlocalized chimeric receptor showed the best performance of THROPPIS. Furthermore, the addition of the helper module dramatically improved the detection sensitivity. In total, 5 peptide-domain interactions were detected successfully, demonstrating the versatility of THROPPIS. In addition, a peptide-domain interaction was detected even when insulin receptor or epidermal growth factor receptor was used as a signaling domain, demonstrating that this PPI detection system can be extended to other receptors.


Subject(s)
Protein Interaction Domains and Motifs/genetics , Protein Interaction Mapping/methods , Receptors, Thrombopoietin , Recombinant Fusion Proteins , Animals , Cell Line , Cell Proliferation/genetics , Mice , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/genetics
2.
J Cell Mol Med ; 25(19): 9073-9083, 2021 10.
Article in English | MEDLINE | ID: mdl-34448528

ABSTRACT

Thrombopoiesis had long been a challenging area of study due to the rarity of megakaryocyte precursors in the bone marrow and the incomplete understanding of its regulatory cytokines. A breakthrough was achieved in the early 1990s with the discovery of the thrombopoietin receptor (TpoR) and its ligand thrombopoietin (TPO). This accelerated research in thrombopoiesis, including the uncovering of the molecular basis of myeloproliferative neoplasms (MPN) and the advent of drugs to treat thrombocytopenic purpura. TpoR mutations affecting its membrane dynamics or transport were increasingly associated with pathologies such as MPN and thrombocytosis. It also became apparent that TpoR affected hematopoietic stem cell (HSC) quiescence while priming hematopoietic stem cells (HSCs) towards the megakaryocyte lineage. Thorough knowledge of TpoR surface localization, dimerization, dynamics and stability is therefore crucial to understanding thrombopoiesis and related pathologies. In this review, we will discuss the mechanisms of TpoR traffic. We will focus on the recent progress in TpoR membrane dynamics and highlight the areas that remain unexplored.


Subject(s)
Receptors, Thrombopoietin/metabolism , Animals , Calreticulin/genetics , Calreticulin/metabolism , Disease Susceptibility , Drug Discovery , Gene Expression Regulation/drug effects , Golgi Apparatus/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Janus Kinase 2/metabolism , Mutation , Protein Binding , Protein Multimerization , Protein Transport , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/genetics , Signal Transduction , Structure-Activity Relationship , TYK2 Kinase/metabolism , Thrombopoietin/metabolism
3.
Int J Mol Sci ; 22(13)2021 Jun 27.
Article in English | MEDLINE | ID: mdl-34199099

ABSTRACT

Eltrombopag is a thrombopoietin receptor (MPL) agonist approved for the treatment of primary immune thrombocytopenia (ITP). Recent evidence shows that some patients may sustain platelet counts following eltrombopag discontinuation. The systemic immunomodulatory response that resolves ITP in some patients could result from an increase in platelet mass, caused either by the direct action of eltrombopag on megakaryocytes through MPL stimulation, or potential MPL-independent actions on other cell types. To uncover the possible mechanisms of action of eltrombopag, in silico analyses were performed, including a systems biology-based approach, a therapeutic performance mapping system, and structural analyses. Through manual curation of the available bibliography, 56 key proteins were identified and integrated into the ITP interactome analysis. Mathematical models (94.92% mean accuracy) were obtained to elucidate potential MPL-dependent pathways in non-megakaryocytic cell subtypes. In addition to the effects on megakaryocytes and platelet numbers, the results were consistent with MPL-mediated effects on other cells, which could involve interferon-gamma, transforming growth factor-beta, peroxisome proliferator-activated receptor-gamma, and forkhead box protein P3 pathways. Structural analyses indicated that effects on three apoptosis-related proteins (BCL2L1, BCL2, BAX) from the Bcl-2 family may be off-target effects of eltrombopag. In conclusion, this study proposes new hypotheses regarding the immunomodulatory functions of eltrombopag in patients with ITP.


Subject(s)
Benzoates/pharmacology , Hydrazines/pharmacology , Immunomodulation/drug effects , Purpura, Thrombocytopenic, Idiopathic/etiology , Purpura, Thrombocytopenic, Idiopathic/metabolism , Pyrazoles/pharmacology , Receptors, Thrombopoietin/antagonists & inhibitors , Benzoates/chemistry , Benzoates/therapeutic use , Biomarkers , Disease Management , Disease Susceptibility , Humans , Hydrazines/chemistry , Hydrazines/therapeutic use , Models, Biological , Models, Molecular , Molecular Targeted Therapy/methods , Protein Interaction Mapping , Protein Interaction Maps , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Pyrazoles/chemistry , Pyrazoles/therapeutic use , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome
4.
Blood ; 137(14): 1920-1931, 2021 04 08.
Article in English | MEDLINE | ID: mdl-33202418

ABSTRACT

Somatic mutations of calreticulin (CALR) have been identified as a main disease driver of myeloproliferative neoplasms, suggesting that development of drugs targeting mutant CALR is of great significance. Site-directed mutagenesis in the N-glycan binding domain (GBD) abolishes the ability of mutant CALR to oncogenically activate the thrombopoietin receptor (MPL). We therefore hypothesized that a small molecule targeting the GBD might inhibit the oncogenicity of the mutant CALR. Using an in silico molecular docking study, we identified candidate binders to the GBD of CALR. Further experimental validation of the hits identified a group of catechols inducing a selective growth inhibitory effect on cells that depend on oncogenic CALR for survival and proliferation. Apoptosis-inducing effects by the compound were significantly higher in the CALR-mutated cells than in CALR wild-type cells. Additionally, knockout or C-terminal truncation of CALR eliminated drug hypersensitivity in CALR-mutated cells. We experimentally confirmed the direct binding of the selected compound to CALR, disruption of the mutant CALR-MPL interaction, inhibition of the JAK2-STAT5 pathway, and reduction at the intracellular level of mutant CALR upon drug treatment. Our data indicate that small molecules targeting the GBD of CALR can selectively kill CALR-mutated cells by disrupting the CALR-MPL interaction and inhibiting oncogenic signaling.


Subject(s)
Calreticulin/metabolism , Hematoxylin/pharmacology , Protein Interaction Maps/drug effects , Receptors, Thrombopoietin/metabolism , Animals , Binding Sites/drug effects , Calreticulin/chemistry , Calreticulin/genetics , Cell Line , Drug Discovery , Humans , Mice , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mutation , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Protein Binding/drug effects , Receptors, Thrombopoietin/chemistry
5.
Science ; 367(6478): 643-652, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32029621

ABSTRACT

Homodimeric class I cytokine receptors are assumed to exist as preformed dimers that are activated by ligand-induced conformational changes. We quantified the dimerization of three prototypic class I cytokine receptors in the plasma membrane of living cells by single-molecule fluorescence microscopy. Spatial and spatiotemporal correlation of individual receptor subunits showed ligand-induced dimerization and revealed that the associated Janus kinase 2 (JAK2) dimerizes through its pseudokinase domain. Oncogenic receptor and hyperactive JAK2 mutants promoted ligand-independent dimerization, highlighting the formation of receptor dimers as the switch responsible for signal activation. Atomistic modeling and molecular dynamics simulations based on a detailed energetic analysis of the interactions involved in dimerization yielded a mechanistic blueprint for homodimeric class I cytokine receptor activation and its dysregulation by individual mutations.


Subject(s)
Carcinogenesis/genetics , Cell Membrane/chemistry , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Protein Multimerization , Receptors, Erythropoietin/chemistry , Receptors, Somatotropin/chemistry , Receptors, Thrombopoietin/chemistry , Amino Acid Substitution/genetics , HeLa Cells , Humans , Janus Kinase 2/antagonists & inhibitors , Ligands , Microscopy, Fluorescence , Models, Molecular , Mutation , Nitriles , Phenylalanine/genetics , Pyrazoles/pharmacology , Pyrimidines , Signal Transduction , Single Molecule Imaging , Valine/genetics
6.
Blood ; 135(4): 287-292, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31697803

ABSTRACT

The single transmembrane domain (TMD) of the human thrombopoietin receptor (TpoR/myeloproliferative leukemia [MPL] protein), encoded by exon 10 of the MPL gene, is a hotspot for somatic mutations associated with myeloproliferative neoplasms (MPNs). Approximately 6% and 14% of JAK2 V617F- essential thrombocythemia and primary myelofibrosis patients, respectively, have "canonical" MPL exon 10 driver mutations W515L/K/R/A or S505N, which generate constitutively active receptors and consequent loss of Tpo dependence. Other "noncanonical" MPL exon 10 mutations have also been identified in patients, both alone and in combination with canonical mutations, but, in almost all cases, their functional consequences and relevance to disease are unknown. Here, we used a deep mutational scanning approach to evaluate all possible single amino acid substitutions in the human TpoR TMD for their ability to confer cytokine-independent growth in Ba/F3 cells. We identified all currently recognized driver mutations and 7 novel mutations that cause constitutive TpoR activation, and a much larger number of second-site mutations that enhance S505N-driven activation. We found examples of both of these categories in published and previously unpublished MPL exon 10 sequencing data from MPN patients, demonstrating that some, if not all, of the new mutations reported here represent likely drivers or modifiers of myeloproliferative disease.


Subject(s)
Amino Acid Substitution , Myeloproliferative Disorders/genetics , Receptors, Thrombopoietin/genetics , Animals , Cell Line , Exons , Humans , Mice , Models, Molecular , Mutation , Protein Domains , Receptors, Thrombopoietin/chemistry
7.
Haematologica ; 104(6): 1112-1123, 2019 06.
Article in English | MEDLINE | ID: mdl-31073079

ABSTRACT

The two thrombopoietin receptor agonists (TPO-RA), eltrombopag and romiplostim, were licensed in the US for treatment of immune thrombocytopenia (ITP) in 2008 and, since then, their use has progressively increased around the world; they are currently used in more than 100 countries. The six largest randomized controlled trials conducted in ITP have used one of these two agents. All studies have demonstrated a platelet response rate between 50-90%, depending on the criteria used, with good safety and tolerability. TPO-RA were shown to be effective in reducing bleeding and the need for concomitant or rescue medication. Many other investigations of their mechanism of effect, prospective and retrospective trials, and studies focusing on toxicity have been performed widening our knowledge of these two agents. Initial concerns on issues such as myelofibrosis have not been confirmed. Only a small number of patients develop moderate-severe reticulin fibrosis and/or collagen fibrosis; however, these are usually reversed after discontinuation of TPO-RA. Studies indicate, however, that TPO-RA may increase the risk of venous thromboembolism. Both TPO-RA are currently approved in patients with chronic ITP aged >1-year who are refractory to at least one other treatment. Eltrombopag has acquired two additional indications: severe aplastic anemia refractory to first-line treatment and hepatitis C patients undergoing treatment with interferon-ribavirin. Despite these wide-ranging studies, important questions still need to be answered. This summary review on TPO-RA will summarize what is known regarding efficacy in ITP, evaluate safety concerns in more depth, and focus on the questions that remain.


Subject(s)
Benzoates/therapeutic use , Hydrazines/therapeutic use , Pyrazoles/therapeutic use , Receptors, Fc/therapeutic use , Receptors, Thrombopoietin/agonists , Recombinant Fusion Proteins/therapeutic use , Thrombopoietin/therapeutic use , Animals , Benzoates/chemistry , Benzoates/pharmacology , Biomarkers , Blood Coagulation/drug effects , Clinical Trials as Topic , Disease Susceptibility , Humans , Hydrazines/chemistry , Hydrazines/pharmacology , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/complications , Purpura, Thrombocytopenic, Idiopathic/drug therapy , Purpura, Thrombocytopenic, Idiopathic/etiology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Receptors, Fc/chemistry , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Signal Transduction/drug effects , Thrombopoietin/chemistry , Thrombopoietin/pharmacology , Treatment Outcome
8.
Blood ; 131(7): 782-786, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29288169

ABSTRACT

Mutations in calreticulin (CALR) are phenotypic drivers in the pathogenesis of myeloproliferative neoplasms. Mechanistic studies have demonstrated that mutant CALR binds to the thrombopoietin receptor MPL, and that the positive electrostatic charge of the mutant CALR C terminus is required for mutant CALR-mediated activation of JAK-STAT signaling. Here we demonstrate that although binding between mutant CALR and MPL is required for mutant CALR to transform hematopoietic cells; binding alone is insufficient for cytokine independent growth. We further show that the threshold of positive charge in the mutant CALR C terminus influences both binding of mutant CALR to MPL and activation of MPL signaling. We find that mutant CALR binds to the extracellular domain of MPL and that 3 tyrosine residues within the intracellular domain of MPL are required to activate signaling. With respect to mutant CALR function, we show that its lectin-dependent function is required for binding to MPL and for cytokine independent growth, whereas its chaperone and polypeptide-binding functionalities are dispensable. Together, our findings provide additional insights into the mechanism of the pathogenic mutant CALR-MPL interaction in myeloproliferative neoplasms.


Subject(s)
Calreticulin/genetics , Calreticulin/metabolism , Myeloproliferative Disorders/genetics , Protein Interaction Domains and Motifs , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/metabolism , Calreticulin/chemistry , Cells, Cultured , HEK293 Cells , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Mutagenesis , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Protein Binding , Protein Interaction Domains and Motifs/genetics , Protein Interaction Maps , Receptors, Thrombopoietin/chemistry , Signal Transduction
9.
Bioorg Med Chem ; 25(3): 1113-1121, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28038942

ABSTRACT

Thrombopoietin (TPO) acts in promoting the proliferation of hematopoietic stem cells and by initiating specific maturation events in megakaryocytes. Now, TPO-mimetic peptides with amino acid sequences unrelated to TPO are of considerable pharmaceutical interest. In the present paper, four new TPO mimetic peptides that bind and activate c-Mpl receptor have been identified, synthesized and tested by Dual-Luciferase reporter gene assay for biological activities. The molecular modeling research was also approached to understand key molecular mechanisms and structural features responsible for peptide binding with c-Mpl receptor. The results presented that three of four mimetic peptides showed significant activities. In addition, the molecular modeling approaches proved hydrophobic interactions were the driven positive forces for binding behavior between peptides and c-Mpl receptor. TPO peptide residues in P7, P13 and P7' positions were identified by the analysis of hydrogen bonds and energy decompositions as the key ones for benefiting better biological activities. Our data suggested the synthesized peptides have considerable potential for the future development of stable and highly active TPO mimetic peptides.


Subject(s)
Models, Molecular , Peptides/chemical synthesis , Peptides/metabolism , Receptors, Thrombopoietin/metabolism , Binding Sites , Humans , Molecular Structure , Peptides/chemistry , Receptors, Thrombopoietin/chemistry , Structure-Activity Relationship
10.
Biochem Biophys Res Commun ; 468(1-2): 262-8, 2015.
Article in English | MEDLINE | ID: mdl-26505790

ABSTRACT

The thrombopoietin receptor, also known as c-Mpl, is a member of the cytokine superfamily, which regulates the differentiation of megakaryocytes and formation of platelets by binding to its ligand, thrombopoietin (TPO), through Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling. The loss-of-function mutations of c-Mpl cause severe thrombocytopenia due to impaired megakaryocytopoiesis, and gain-of-function mutations cause thrombocythemia. c-Mpl contains two Trp-Ser-Xaa-Trp-Ser (Xaa represents any amino acids) sequences, which are characteristic sequences of type I cytokine receptors, corresponding to C-mannosylation consensus sequences: Trp-Xaa-Xaa-Trp/Cys. C-mannosylation is a post-translational modification of tryptophan residue in which one mannose is attached to the first tryptophan residue in the consensus sequence via C-C linkage. Although c-Mpl contains some C-mannosylation sequences, whether c-Mpl is C-mannosylated or not has been uninvestigated. We identified that c-Mpl is C-mannosylated not only at Trp(269) and Trp(474), which are putative C-mannosylation site, but also at Trp(272), Trp(416), and Trp(477). Using C-mannosylation defective mutant of c-Mpl, the C-mannosylated tryptophan residues at four sites (Trp(269), Trp(272), Trp(474), and Trp(477)) are essential for c-Mpl-mediated JAK-STAT signaling. Our findings suggested that C-mannosylation of c-Mpl is a possible therapeutic target for platelet disorders.


Subject(s)
Receptors, Thrombopoietin/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Thrombopoietin/metabolism , Tryptophan/analogs & derivatives , Amino Acid Sequence , Cell Line , Humans , Janus Kinases/metabolism , Molecular Sequence Data , Receptors, Thrombopoietin/chemistry , Tryptophan/analysis , Tryptophan/metabolism
11.
Blood ; 125(6): 941-8, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25468569

ABSTRACT

Thrombopoietin (Thpo) signaling through the c-Mpl receptor promotes either quiescence or proliferation of hematopoietic stem cells (HSCs) in a concentration-dependent manner; however, in vivo Thpo serum levels are responsive to platelet mass rather than HSC demands, suggesting additional regulation exists. Ott1 (Rbm15), a spliceosomal component originally identified as a fusion partner in t(1;22)-associated acute megakaryocytic leukemia, is also essential for maintaining HSC quiescence under stress. Ott1 controls the alternative splicing of a dominant negative isoform, Mpl-TR, capable of inhibiting HSC engraftment and attenuating Thpo signaling. Ott1, which associates with Hdac3 and the histone methyltransferase, Setd1b, binds to both c-Mpl RNA and chromatin and regulates H4 acetylation and H3K4me3 marks. Histone deacetylase or histone methyltransferase inhibition also increases Mpl-TR levels, suggesting that Ott1 uses an underlying epigenetic mechanism to control alternative splicing of c-Mpl. Manipulation of Ott1-dependent alternative splicing may therefore provide a novel pharmacologic avenue for regulating HSC quiescence and proliferation in response to Thpo.


Subject(s)
Alternative Splicing , Hematopoietic Stem Cells/metabolism , RNA-Binding Proteins/metabolism , Receptors, Thrombopoietin/genetics , Thrombopoietin/metabolism , Animals , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, Knockout , NIH 3T3 Cells , Protein Isoforms/chemistry , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Receptors, Thrombopoietin/chemistry , Signal Transduction
12.
Mol Cell Biochem ; 391(1-2): 85-94, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24615392

ABSTRACT

Thrombopoietin (TPO) can regulate hematopoiesis and megakaryopoiesis via activation of its receptor, c-Mpl, and multiple downstream signal transduction pathways. Using the cytoplasmic domain of Mpl as bait, we performed yeast two-hybrid screening, and found that the protein Atp5d might associate with Mpl. Atp5d is known as the δ subunit of mitochondrial ATP synthase, but little is known about the function of dissociative Atp5d. The interaction between Mpl and Atp5d was confirmed by the yeast two-hybrid system, mammalian two-hybrid assay, pull-down experiment, and co-immunoprecipitation study in vivo and in vitro. An additional immunofluorescence assay showed that the two proteins can colocalize along the plasma membrane in the cytoplasm. Using the yeast two-hybrid system, we tested a series of cytoplasmic truncated mutations for their ability to bind Atp5d and found an association between Atp5d and the Aa98-113 domain of Mpl. The dissociation of Atp5d from Mpl after TPO stimulation suggests that Atp5d may be a new component of TPO signaling.


Subject(s)
Proton-Translocating ATPases/metabolism , Receptors, Thrombopoietin/metabolism , Animals , Cell Line , Humans , Intracellular Space/metabolism , Mice , Mitochondrial Proton-Translocating ATPases , Protein Binding/drug effects , Protein Interaction Domains and Motifs , Protein Interaction Maps/drug effects , Protein Structure, Tertiary , Receptors, Thrombopoietin/chemistry , Reproducibility of Results , Thrombopoietin/pharmacology , Two-Hybrid System Techniques
13.
Stem Cells ; 32(6): 1503-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24677652

ABSTRACT

Unlimited self renewal capacity and differentiation potential make human pluripotent stem cells (PSC) a promising source for the ex vivo manufacture of red blood cells (RBCs) for safe transfusion. Current methods to induce erythropoiesis from PSC suffer from low yields of RBCs, most of which are immature and contain embryonic and fetal rather than adult hemoglobins. We have previously shown that homodimerization of the intracellular component of MPL (ic-MPL) induces erythropoiesis from human cord blood progenitors. The goal of this study was to investigate the potential of ic-MPL dimerization to induce erythropoiesis from human embryonic stem cells (hESCs) and to identify the signaling pathways activated by this strategy. We present here the evidence that ic-MPL dimerization induces erythropoietin (EPO)-independent erythroid differentiation from hESC by inducing the generation of erythroid progenitors and by promoting more efficient erythroid maturation with increased RBC enucleation as well as increased gamma:epsilon globin ratio and production of beta-globin protein. ic-MPL dimerization is significantly more potent than EPO in inducing erythropoiesis, and its effect is additive to EPO. Signaling studies show that dimerization of ic-MPL, unlike stimulation of the wild type MPL receptor, activates AKT in the absence of JAK2/STAT5 signaling. AKT activation upregulates GATA-1 and FOXO3 transcriptional pathways with resulting inhibition of apoptosis, modulation of cell cycle, and enhanced maturation of erythroid cells. These findings open up potential new targets for the generation of therapeutically relevant RBC products from hPSC.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Erythropoiesis , Erythropoietin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Cell Cycle , Cell Differentiation , Cell Line , Cell Proliferation , Cell Survival , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Humans , Megakaryocytes/cytology , Megakaryocytes/metabolism , Protein Multimerization , Protein Structure, Tertiary , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/metabolism
14.
Biotechnol Prog ; 30(2): 411-7, 2014.
Article in English | MEDLINE | ID: mdl-24375974

ABSTRACT

Since cell migration plays critical roles in development and homeostasis of the body, artificial control of cell migration would be promising for the treatment of various diseases related to migration. To this end, we previously developed single-chain Fv (scFv)/receptor chimeras, named signalobodies, which can control cell fates via a specific antigen that is different from natural cytokines. Although a conventional chemotaxis chamber assay revealed that several signalobodies based on receptor tyrosine kinases transduced antigen-dependent migration signals, we have never performed direct observation of the cells to obtain more information on overall properties of cell motility and migration. In this study, we utilized murine pro-B Ba/F3 cells expressing either a scFv-Fms or scFv-Mpl signalobody, and compared their migratory characteristics. We employed a lipid-polyethylene glycol conjugate to softly immobilize the suspension cells on a slide, which facilitated direct observation of chemokinetic activity of the cells. Consequently, both cells markedly exhibited chemokinesis in response to a specific antigen. In addition, the cells were subjected to a stable antigen-concentration gradient to observe horizontal directional cell migration in real time. The results showed that the cells expressing scFv-Fms underwent directional migration toward a positive antigen-concentration gradient. Taken together, we successfully demonstrated antigen-responsive regulation of cell motility and migration via the signalobodies.


Subject(s)
Cell Movement/physiology , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Thrombopoietin/metabolism , Signal Transduction/physiology , Single-Chain Antibodies/metabolism , Animals , Antigens/metabolism , Cell Line , Fluorescein/chemistry , Fluorescein/metabolism , Mice , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Receptors, Thrombopoietin/chemistry , Single-Chain Antibodies/chemistry
15.
Int J Biochem Cell Biol ; 45(12): 2852-63, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24144576

ABSTRACT

The thrombopoietin receptor is a crucial element in thrombopoietin-initiated signaling pathways, which stimulates the differentiation of normal hematopoietic progenitor cells, the maturation of megakaryocytes, and the generation of platelets. In this study, we identified a novel activating variant of thrombopoietin receptor, termed Mpl-D, in human megakaryoblastic leukemia Dami cells and demonstrated that the binding affinity of the Mpl-D receptor for thrombopoietin is enhanced. Cell cycle analysis revealed that in the presence of thrombopoietin, most Mpl-D expressing NIH3T3 (NIH3T3/Mpl-D) cells were prevalent in G1 phase while the S and G2/M populations were less frequently observed. Unexpectedly, thrombopoietin induced strong and prolonged ERK1/2 signaling in NIH3T3/Mpl-D cells compared with its receptor wild-type expressing NIH3T3 (NIH3T3/Mpl-F) cells. Further analysis of the mRNA levels of cyclin D1/D2 in NIH3T3/Mpl-D cells demonstrated markedly down-regulated expression compared to NIH3T3/Mpl-F cells in the presence of thrombopoietin. Thus, the prolonged activation of ERK1/2 by Mpl-D might lead to G1 cell cycle arrest through a profound reduction of cyclin D1/D2 in order to support cell survival without proliferation. We also provided tertiary structural basis for the Mpl-D and thrombopoietin interaction, which might provide insights into how Mpl-D effectively increases binding to thrombopoietin and significantly contributes to its specific signaling pathway. These results suggest a new paradigm for the regulation of cytokine receptor expression and function through the alternative splicing variant of Mpl in Dami cells, which may play a role in the pathogenesis of megakaryoblastic leukemia.


Subject(s)
MAP Kinase Signaling System/physiology , Receptors, Thrombopoietin/metabolism , Thrombopoietin/metabolism , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Humans , MAP Kinase Signaling System/drug effects , Mice , Molecular Sequence Data , NIH 3T3 Cells , Protein Conformation , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/genetics , Signal Transduction , Thrombopoietin/genetics , Thrombopoietin/pharmacology
16.
Proc Natl Acad Sci U S A ; 110(7): 2540-5, 2013 Feb 12.
Article in English | MEDLINE | ID: mdl-23359689

ABSTRACT

Dimerization of single-pass membrane receptors is essential for activation. In the human thrombopoietin receptor (TpoR), a unique amphipathic RWQFP motif separates the transmembrane (TM) and intracellular domains. Using a combination of mutagenesis, spectroscopy, and biochemical assays, we show that W515 of this motif impairs dimerization of the upstream TpoR TM helix. TpoR is unusual in that a specific residue is required for this inhibitory function, which prevents receptor self-activation. Mutations as diverse as W515K and W515L cause oncogenic activation of TpoR and lead to human myeloproliferative neoplasms. Two lines of evidence support a general mechanism in which W515 at the intracellular juxtamembrane boundary inhibits dimerization of the TpoR TM helix by increasing the helix tilt angle relative to the membrane bilayer normal, which prevents the formation of stabilizing TM dimer contacts. First, measurements using polarized infrared spectroscopy show that the isolated TM domain of the active W515K mutant has a helix tilt angle closer to the bilayer normal than that of the wild-type receptor. Second, we identify second-site R514W and Q516W mutations that reverse dimerization and tilt angle changes induced by the W515K and W515L mutations. The second-site mutations prevent constitutive activation of TpoR W515K/L, while preserving ligand-induced signaling. The ability of tryptophan to influence the angle and dimerization of the TM helix in wild-type TpoR and in the second-site revertants is likely associated with its strong preference to be buried in the headgroup region of membrane bilayers.


Subject(s)
Models, Molecular , Protein Conformation , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/metabolism , Tryptophan/metabolism , Amino Acid Motifs/genetics , Animals , Base Sequence , Blotting, Western , Cell Line , Cloning, Molecular , Dimerization , Flow Cytometry , Genetic Complementation Test , Humans , Luciferases , Magnetic Resonance Spectroscopy , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Receptors, Thrombopoietin/genetics , Sequence Analysis, DNA , Spectrophotometry, Infrared , Ultracentrifugation
17.
Am J Hematol ; 87(5): 532-4, 2012 May.
Article in English | MEDLINE | ID: mdl-22389068

ABSTRACT

Essential thrombocythemia (ET) is a rare type of myeloproliferative neoplasm characterized by clonal expansion of the megakaryocyte and platelet lineage. Here, we describe a novel mutation (Y252H) in the thrombopoietin (TPO) receptor, or MPL, in a JAK2 mutation-negative ET patient. The bone marrow examination revealed increased numbers of dysmorphic megakaryocytes with focal clustering. The x-inactivation pattern suggested clonal expansion of hematopoietic cells in the bone marrow. Furthermore, we found that the patient's bone marrow cells were hypersensitive to TPO in generating megakaryocyte colonies in vitro. More importantly, we demonstrated that this MPL Y252H mutant confers increased TPO/MPL-mediated cell growth and increased cell survival upon cytokine withdrawal in BaF3 cells, indicating it is a disease-driving mutation and may contribute to the development of ET in vivo. In summary, this is the first report describing a mutation in the extracellular domain of MPL underlying ET.


Subject(s)
Mutation, Missense , Point Mutation , Receptors, Thrombopoietin/genetics , Thrombocythemia, Essential/genetics , Thrombopoietin/pharmacology , Amino Acid Substitution , Animals , Bone Marrow/pathology , Cell Line, Transformed/drug effects , Child, Preschool , Clone Cells/pathology , DNA Mutational Analysis , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Janus Kinase 2/genetics , Megakaryocytes/pathology , Mice , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/physiology , Thrombocythemia, Essential/pathology , X Chromosome Inactivation
19.
EMBO J ; 30(21): 4398-413, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21892137

ABSTRACT

Ligand binding to the thrombopoietin receptor is thought to stabilize an active receptor dimer that regulates megakaryocyte differentiation and platelet formation, as well as haematopoietic stem cell renewal. By fusing a dimeric coiled coil in all seven possible orientations to the thrombopoietin receptor transmembrane (TM)-cytoplasmic domains, we show that specific biological effects and in vivo phenotypes are imparted by distinct dimeric orientations, which can be visualized by cysteine mutagenesis and crosslinking. Using functional assays and computational searches, we identify one orientation that represents the inactive dimeric state and another similar to a physiologically activated receptor. Several other dimeric orientations are identified that induce proliferation and in vivo myeloproliferative and myelodysplastic disorders, indicating the receptor can signal from several dimeric interfaces. The set of dimeric thrombopoietin receptors with different TM orientations may offer new insights into the activation of distinct signalling pathways by a single receptor and suggests that subtle differences in cytokine receptor dimerization provide a new layer of signalling regulation that is relevant for disease.


Subject(s)
Protein Multimerization/physiology , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Protein Interaction Maps , Protein Multimerization/genetics , Receptors, Thrombopoietin/genetics , Receptors, Thrombopoietin/physiology , Signal Transduction/physiology , Stereoisomerism
20.
PLoS One ; 6(8): e23396, 2011.
Article in English | MEDLINE | ID: mdl-21858098

ABSTRACT

BACKGROUND: Mutations in the thrombopoietin receptor (MPL) may activate relevant pathways and lead to chronic myeloproliferative neoplasms (MPNs). The mechanisms of MPL activation remain elusive because of a lack of experimental structures. Modern computational biology techniques were utilized to explore the mechanisms of MPL protein activation due to various mutations. RESULTS: Transmembrane (TM) domain predictions, homology modeling, ab initio protein structure prediction, and molecular dynamics (MD) simulations were used to build structural dynamic models of wild-type and four clinically observed mutants of MPL. The simulation results suggest that S505 and W515 are important in keeping the TM domain in its correct position within the membrane. Mutations at either of these two positions cause movement of the TM domain, altering the conformation of the nearby intracellular domain in unexpected ways, and may cause the unwanted constitutive activation of MPL's kinase partner, JAK2. CONCLUSIONS: Our findings represent the first full-scale molecular dynamics simulations of the wild-type and clinically observed mutants of the MPL protein, a critical element of the MPL-JAK2-STAT signaling pathway. In contrast to usual explanations for the activation mechanism that are based on the relative translational movement between rigid domains of MPL, our results suggest that mutations within the TM region could result in conformational changes including tilt and rotation (azimuthal) angles along the membrane axis. Such changes may significantly alter the conformation of the adjacent and intrinsically flexible intracellular domain. Hence, caution should be exercised when interpreting experimental evidence based on rigid models of cytokine receptors or similar systems.


Subject(s)
Models, Molecular , Mutation , Protein Structure, Tertiary , Receptors, Thrombopoietin/chemistry , Receptors, Thrombopoietin/genetics , Amino Acid Substitution , Computational Biology/methods , Humans , Janus Kinase 2/chemistry , Janus Kinase 2/metabolism , Models, Biological , Molecular Conformation , Molecular Dynamics Simulation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Poly-ADP-Ribose Binding Proteins , Protein Inhibitors of Activated STAT , Receptors, Thrombopoietin/metabolism , STAT Transcription Factors/chemistry , STAT Transcription Factors/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...