Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Publication year range
1.
Blood ; 134(26): 2388-2398, 2019 12 26.
Article in English | MEDLINE | ID: mdl-31697804

ABSTRACT

The V617F mutation in the JH2 domain of Janus kinase 2 (JAK2) is an oncogenic driver in several myeloproliferative neoplasms (MPNs), including essential thrombocythemia, myelofibrosis, and polycythemia vera (PV). Other mutations in JAK2 have been identified in MPNs, most notably exon 12 mutations in PV. Here, we describe a novel recurrent mutation characterized by a common 4-amino-acid deletion and variable 1-amino-acid insertion (Leu583-Ala586DelInsSer/Gln/Pro) within the JH2 domain of JAK2. All 4 affected patients had eosinophilia, and both patients with Leu583-Ala586DelInsSer fulfilled diagnostic criteria of both PV and chronic eosinophilic leukemia (CEL). Computational and functional studies revealed that Leu583-Ala586DelInsSer (herein referred to as JAK2ex13InDel) deregulates JAK2 through a mechanism similar to JAK2V617F, activates signal transducer and activator of transcription 5 and extracellular signal-regulated kinase, and transforms parental Ba/F3 cells to growth factor independence. In contrast to JAK2V617F, JAK2ex13InDel does not require an exogenous homodimeric type 1 cytokine receptor to transform Ba/F3 cells and is capable of activating ß common chain family cytokine receptor (interleukin-3 receptor [IL-3R], IL-5R, and granulocyte-macrophage colony stimulating factor receptor) signaling in the absence of ligand, with the maximum effect observed for IL-5R, consistent with the clinical phenotype of eosinophilia. Recognizing this new PV/CEL-overlap MPN has significant clinical implications, as both PV and CEL patients are at high risk for thrombosis, and concomitant cytoreduction of red cells, neutrophils, and eosinophils may be required for prevention of thromboembolic events. Targeted next-generation sequencing for genes recurrently mutated in myeloid malignancies in patients with unexplained eosinophilia may reveal additional cases of Leu583-Ala586DelInsSer/Gln/Pro, allowing for complete characterization of this unique MPN.


Subject(s)
B-Lymphocytes/pathology , Cell Transformation, Neoplastic/pathology , Hypereosinophilic Syndrome/pathology , INDEL Mutation , Janus Kinase 2/genetics , Leukemia/pathology , Polycythemia Vera/pathology , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Animals , B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cells, Cultured , Clonal Evolution , Female , Humans , Hypereosinophilic Syndrome/genetics , Hypereosinophilic Syndrome/metabolism , Janus Kinase 2/metabolism , Leukemia/genetics , Leukemia/metabolism , Male , Mice , Oncogenes , Polycythemia Vera/genetics , Polycythemia Vera/metabolism
2.
Mol Carcinog ; 59(1): 87-103, 2020 01.
Article in English | MEDLINE | ID: mdl-31691359

ABSTRACT

Hypereosinophilia (HE) is caused by a variety of disorders, ranging from parasite infections to autoimmune diseases and cancer. Only a small proportion of HE cases are clonal malignancies, and one of these, the group of eosinophilia-associated tyrosine kinase fusion-driven neoplasms, is sensitive to tyrosine kinase inhibitors, while most subtypes lack specific treatment. Eosinophil functions are highly dependent on actin polymerization, promoting priming, shape change, and infiltration of inflamed tissues. Therefore, we investigated the role of the actin-binding protein lymphocyte cytosolic protein 1 (LCP1) in malignant and nonmalignant eosinophil differentiation. We use the protein kinase C-ß (PKCß) selective inhibitor enzastaurin (Enza) to dephosphorylate and inactivate LCP1 in FIP1L1-platelet-derived growth factor receptor α (PDGFRA)-positive Eol-1 cells, and this was associated with reduced proliferation, metabolic activity, and colony formation as well as enhanced apoptosis and impaired migration. While Enza did not alter FIP1L1-PDGFRA-induced signal transducer and activator of transcription 3 (STAT3), STAT5, and ERK1/2 phosphorylation, it inhibited STAT1Tyr701 and AKTSer473 (but not AKTThr308 ) phosphorylation, and short hairpin RNA knockdown experiments confirmed that this process was mediated by LCP1 and associated mammalian target of rapamycin complex 2 (mTORC2) activity loss. Homeobox protein HoxB8 immortalized murine bone marrow cells showed impaired eosinophilic differentiation upon Enza treatment or LCP1 knockdown. Furthermore, Enza treatment of primary HE samples reduced eosinophil differentiation and survival. In conclusion, our data show that HE involves active LCP1, which interacts with mTOR and triggers mTORC2 activity, and that the PKCß inhibitor Enza as well as targeting of LCP1 may provide a novel treatment approach to hypereosinophilic disorders.


Subject(s)
Hypereosinophilic Syndrome/drug therapy , Indoles/pharmacology , Mechanistic Target of Rapamycin Complex 2/metabolism , Microfilament Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Adult , Aged , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Hypereosinophilic Syndrome/metabolism , Indoles/therapeutic use , Male , Middle Aged , Protein Kinase Inhibitors/therapeutic use
3.
Am J Physiol Heart Circ Physiol ; 317(2): H405-H414, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31199184

ABSTRACT

Hypereosinophilic syndrome is characterized by sustained and marked eosinophilia leading to tissue damage and organ dysfunction. Morbidity and mortality occur primarily due to cardiac and thromboembolic complications. Understanding the cause and mechanism of disease would aid in the development of targeted therapies with greater efficacy and fewer side effects. We discovered a spontaneous mouse mutant in our colony with a hypereosinophilic phenotype. Mice develop peripheral blood eosinophilia; infiltration of lungs, spleen, and heart by eosinophils; and extensive myocardial damage and remodeling. This ultimately leads to heart failure and premature death. Histopathological assessment of the hearts revealed a robust inflammatory infiltrate composed primarily of eosinophils and B-lymphocytes, associated with myocardial damage and replacement fibrosis, consistent with eosinophilic myocarditis. In many cases, hearts showed dilatation and thinning of the right ventricular wall, suggestive of an inflammatory dilated cardiomyopathy. Most mice showed atrial thrombi, which often filled the chamber. Protein expression analysis revealed overexpression of chemokines and cytokines involved in innate and adaptive immunity including IL-4, eotaxin, and RANTES. Disease could be transferred to wild-type mice by adoptive transfer of splenocytes from affected mice, suggesting a role for the immune system. In summary, the pathologies observed in the mutant lines are reminiscent of those seen in patients with hypereosinophilia, where cardiac-related morbidities, like congestive heart failure and thrombi, are the most common causes of death. As such, our model provides an opportunity to test mechanistic hypotheses and develop targeted therapies.NEW & NOTEWORTHY This article describes a new model of heart disease in hypereosinophilia. The model developed as a spontaneous mouse mutant in the colony and is characterized by peripheral blood eosinophilia and infiltration of lungs, spleen, and heart by eosinophils. In the heart, there is extensive myocardial damage, remodeling, fibrosis, and thrombosis, leading to heart failure and death. The immune microenvironment is one of increased innate and adaptive immunity, including Th1 and Th2 cytokines/chemokines. Finally, adoptive transfer of splenocytes transfers disease to recipient mice. In summary, this model provides an opportunity to test mechanistic hypotheses and develop targeted therapies for this rare but devastating disease.


Subject(s)
Cardiomyopathy, Dilated/etiology , Heart Failure/etiology , Hypereosinophilic Syndrome/complications , Myocarditis/etiology , Myocardium , Adaptive Immunity , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Eosinophils/immunology , Eosinophils/metabolism , Fibrosis , Genetic Predisposition to Disease , Heart Failure/immunology , Heart Failure/metabolism , Heart Failure/pathology , Hypereosinophilic Syndrome/immunology , Hypereosinophilic Syndrome/metabolism , Hypereosinophilic Syndrome/pathology , Immunity, Innate , Mice, Mutant Strains , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology , Phenotype , Signal Transduction , Time Factors , Ventricular Remodeling
4.
Cancer Sci ; 108(2): 200-207, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27960034

ABSTRACT

Fusion tyrosine kinases play a crucial role in the development of hematological malignancies. FIP1L1-PDGFRA is a leukemogenic fusion kinase that causes chronic eosinophilic leukemia. As a constitutively active kinase, FIP1L1-PDGFRA stimulates downstream signaling molecules, leading to cellular proliferation and the generation of an anti-apoptotic state. Contribution of the N-terminal FIP1L1 portion is necessary for FIP1L1-PDGFRA to exert its full transforming activity, but the underlying mechanisms have not been fully characterized. We identified PIAS1 as a FIP1L1-PDGFRA association molecule by yeast two-hybrid screening. Our analyses indicate that the FIP1L1 portion of FIP1L1-PDGFRA is required for efficient association with PIAS1. As a consequence of the association, FIP1L1-PDGFRA phosphorylates PIAS1. Moreover, the kinase activity of FIP1L1-PDGFRA stabilizes PIAS1. Therefore, PIAS1 is one of the downstream targets of FIP1L1-PDGFRA. Moreover, we found that PIAS1, as a SUMO E3 ligase, sumoylates and stabilizes FIP1L1-PDGFRA. In addition, suppression of PIAS1 activity by a knockdown experiment resulted in destabilization of FIP1L1-PDGFRA. Therefore, FIP1L1-PDGFRA and PIAS1 form a positive cross-talk through their enzymatic activities. Suppression of sumoylation by ginkgolic acid, a small molecule compound inhibiting a SUMO E1-activating enzyme, also destabilizes FIP1L1-PDGFRA, and while the tyrosine kinase inhibitor imatinib suppresses FIP1L1-PDGFRA-dependent cell growth, ginkgolic acid or siRNA of PIAS1 has a synergistic effect with imatinib. In conclusion, our results suggest that sumoylation by PIAS1 is a potential target in the treatment of FIP1L1-PDGFRA-positive chronic eosinophilic leukemia.


Subject(s)
Cell Nucleus/metabolism , Oncogene Proteins, Fusion/metabolism , Protein Inhibitors of Activated STAT/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , STAT1 Transcription Factor/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Apoptosis , HEK293 Cells , Humans , Hypereosinophilic Syndrome/drug therapy , Hypereosinophilic Syndrome/metabolism , Imatinib Mesylate/therapeutic use , Immunoblotting , Immunoprecipitation , Oncogene Proteins, Fusion/chemistry , Protein Inhibitors of Activated STAT/chemistry , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/chemistry , STAT1 Transcription Factor/chemistry , Signal Transduction , Sumoylation , Transfection/methods , mRNA Cleavage and Polyadenylation Factors/chemistry
6.
Haematologica ; 100(8): 1086-95, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25682606

ABSTRACT

The CD3(-)CD4(+) lymphoid variant of hypereosinophilic syndrome is characterized by hypereosinophilia and clonal circulating CD3(-)CD4(+) T cells. Peripheral T-cell lymphoma has been described during this disease course, and we observed in our cohort of 23 patients 2 cases of angio-immunoblastic T-cell lymphoma. We focus here on histopathological (n=12 patients) and immunophenotypic (n=15) characteristics of CD3(-)CD4(+) lymphoid variant of hypereosinophilic syndrome. Atypical CD4(+) T cells lymphoid infiltrates were found in 10 of 12 CD3(-)CD4(+) L-HES patients, in lymph nodes (n=4 of 4 patients), in skin (n=9 of 9) and other extra-nodal tissues (gut, lacrymal gland, synovium). Lymph nodes displayed infiltrates limited to the interfollicular areas or even an effacement of nodal architecture, associated with proliferation of arborizing high endothelial venules and increased follicular dendritic cell meshwork. Analysis of 2 fresh skin samples confirmed the presence of CD3(-)CD4(+) T cells. Clonal T cells were detected in at least one tissue in 8 patients, including lymph nodes (n=4 of 4): the same clonal T cells were detected in blood and in at least one biopsy, with a maximum delay of 23 years between samples. In the majority of cases, circulating CD3(-)CD4(+) T cells were CD2(hi) (n=9 of 14), CD5(hi) (n=12 of 14), and CD7(-)(n=4 of 14) or CD7(low) (n=10 of 14). Angio-immunoblastic T-cell lymphoma can also present with CD3(-)CD4(+) T cells; despite other common histopathological and immunophenotypic features, CD10 expression and follicular helper T-cell markers were not detected in lymphoid variant of hypereosinophilic syndrome patients, except in both patients who developed angio-immunoblastic T-cell lymphoma, and only at T-cell lymphoma diagnosis. Taken together, persistence of tissular clonal T cells and histopathological features define CD3(-)CD4(+) lymphoid variant of hypereosinophilic syndrome as a peripheral indolent clonal T-cell lymphoproliferative disorder, which should not be confused with angio-immunoblastic T-cell lymphoma.


Subject(s)
CD3 Complex/metabolism , CD4 Antigens/metabolism , Clonal Evolution , Hypereosinophilic Syndrome/metabolism , Hypereosinophilic Syndrome/pathology , Immunophenotyping , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Adolescent , Adult , Aged , Bone Marrow/metabolism , Bone Marrow/pathology , Diagnosis, Differential , Female , Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor , Humans , Hypereosinophilic Syndrome/diagnosis , Hypereosinophilic Syndrome/therapy , Immunohistochemistry , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphoma, T-Cell, Peripheral/diagnosis , Lymphoma, T-Cell, Peripheral/metabolism , Lymphoma, T-Cell, Peripheral/pathology , Male , Middle Aged , Skin/metabolism , Skin/pathology , Young Adult
7.
Eksp Klin Gastroenterol ; (9): 70-4, 2015.
Article in Russian | MEDLINE | ID: mdl-26931014

ABSTRACT

Pathological eosinophilic infiltration of the various parts of the gastrointestinal tract (GIT) is a marker of a wide range of diseases, combined into three groups: eosinophil-associated gastrointestinal diseases (primary gastrointestinal eosinophilia), gastrointestinal disorders within the hypereosinophilic syndrome, gastrointestinal eosinophilia, developing, for obvious reasons. The article provides an overview of foreing and domestic literature about ethiopathogenesis, diagnosis and treatment methods of these three groups of gastrointestinal eosinophilia.


Subject(s)
Eosinophilic Esophagitis , Gastrointestinal Tract , Hypereosinophilic Syndrome , Animals , Eosinophilic Esophagitis/metabolism , Eosinophilic Esophagitis/pathology , Eosinophilic Esophagitis/physiopathology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/pathology , Gastrointestinal Tract/physiopathology , Humans , Hypereosinophilic Syndrome/metabolism , Hypereosinophilic Syndrome/pathology , Hypereosinophilic Syndrome/physiopathology
8.
Mol Cancer ; 13: 17, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24472312

ABSTRACT

BACKGROUND: T674I FIP1L1-PDGFRα in a subset of chronic eosinophilic leukemia (CEL) is a gatekeeper mutation that is resistant to many tyrosine kinase inhibitors (TKIs) (e.g., imatinib, nilotinib and dasatinib), similar to T315I Bcr-Abl. Therefore, novel TKIs effective against T674I FIP1L1-PDGFRα are needed. Ponatinib (AP24534) is a novel orally bioavailable TKI against T315I Bcr-Abl, but it is not clear whether ponatinib is effective against T674I FIP1L1-PDGFRα. The purpose of this study was to examine the effect of ponatinib on T674I FIP1L1-PDGFRα. METHODS: Molecular docking analysis in silico was performed. The effects of ponatinib on PDGFRα signaling pathways, apoptosis and cell cycling were examined in EOL-1, BaF3 cells expressing either wild type (WT) or T674I FIP1L1-PDGFRα. The in vivo antitumor activity of ponatinib was evaluated with xenografted BaF3-T674I FIP1L1-PDGFRα cells in nude mice models. RESULTS: Molecular docking analysis revealed that ponatinib could bind to the DFG (Asp-Phe-Gly)-out state of T674I PDGFRα. Ponatinib potently inhibited the phosphorylation of WT and T674I FIP1L1-PDGFRα and their downstream signaling molecules (e.g., Stat3, Stat5). Ponatinib strikingly inhibited the growth of both WT and T674I FIP1L1-PDGFRα-carrying CEL cells (IC50: 0.004-2.5 nM). It induced apoptosis in CEL cells with caspase-3-dependent cleavage of Mcl-1, and inhibited tyrosine phosphorylation of ß-catenin to decrease its stability and pro-survival functions. In vivo, ponatinib abrogated the growth of xenografted BaF3-T674I FIP1L1-PDGFRα cells in nude mice. CONCLUSIONS: Ponatinib is a pan-FIP1L1-PDGFRα inhibitor, and clinical trials are warranted to investigate its efficacy in imatinib-resistant CEL.


Subject(s)
Antineoplastic Agents/pharmacology , Hypereosinophilic Syndrome/metabolism , Imidazoles/pharmacology , Pyridazines/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Humans , Hypereosinophilic Syndrome/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Molecular Docking Simulation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Oncogene Proteins, Fusion/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Transfection , Xenograft Model Antitumor Assays , beta Catenin/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics
10.
J Immunol ; 188(9): 4602-10, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22474019

ABSTRACT

Immune cell-mediated tissue injury is a common feature of different inflammatory diseases, yet the pathogenetic mechanisms and cell types involved vary significantly. Hypereosinophilic syndrome (HES) represents a group of inflammatory diseases that is characterized by increased numbers of pathogenic eosinophilic granulocytes in the peripheral blood and diverse organs. On the basis of clinical and laboratory findings, various forms of HES have been defined, yet the molecular mechanism and potential signaling pathways that drive eosinophil expansion remain largely unknown. In this study, we show that mice deficient of the serine/threonine-specific protein kinase NF-κB-inducing kinase (NIK) develop a HES-like disease, reflected by progressive blood and tissue eosinophilia, tissue injury, and premature death at around 25-30 wk of age. Similar to the lymphocytic form of HES, CD4(+) T cells from NIK-deficient mice express increased levels of Th2-associated cytokines, and eosinophilia and survival of NIK-deficient mice could be prevented completely by genetic ablation of CD4(+) T cells. Experiments based on bone marrow chimeric mice, however, demonstrated that inflammation in NIK-deficient mice depended on radiation-resistant tissues, implicating that NIK-deficient immune cells mediate inflammation in a nonautonomous manner. Surprisingly, disease development was independent of NIK's known function as an IκB kinase α (IKKα) kinase, because mice carrying a mutation in the activation loop of IKKα, which is phosphorylated by NIK, did not develop inflammatory disease. Our data show that NIK activity in nonhematopoietic cells controls Th2 cell development and prevents eosinophil-driven inflammatory disease, most likely using a signaling pathway that operates independent of the known NIK substrate IKKα.


Subject(s)
Hypereosinophilic Syndrome/immunology , I-kappa B Kinase/immunology , Protein Serine-Threonine Kinases/immunology , Animals , Enzyme Activation/genetics , Enzyme Activation/immunology , Hypereosinophilic Syndrome/genetics , Hypereosinophilic Syndrome/metabolism , Hypereosinophilic Syndrome/pathology , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Knockout , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Th2 Cells/immunology , Th2 Cells/metabolism , Th2 Cells/pathology , NF-kappaB-Inducing Kinase
11.
Curr Opin Hematol ; 20(2): 157-62, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23385615

ABSTRACT

PURPOSE OF REVIEW: In 2012, idiopathic hypereosinophilic syndrome (HES) is still the prevalent diagnosis in patients with persistent eosinophilia, in which a primary or secondary cause of eosinophilia has not been identified. HES is considered a provisional diagnosis until a primary or secondary cause of hypereosinophilia is established. The discovery of imatinib-sensitive fusion proteins in a subset of patients with hypereosinophilia has changed the way we approach the diagnosis and treatment of eosinophilic myeloid neoplasms [eosinophilic myeloproliferative neoplasms (MPNs)]. Despite the recent diagnostic developments, diagnosis of hypereosinophilic MPN is only made in 10-20% of patients with persistent primary hypereosinophilia. RECENT FINDINGS: In 2008 the World Health Organization (WHO) established a semi-molecular classification of hypereosinophilic MPNs. The discovery of PDGFRA, PDGFRB, FGFR1, JAK-2, and FLT3 fusion proteins in patients with eosinophilic MPNs provide opportunities for targeted therapy. Patients with hypereosinophilic MPNs associated with PDGFRA and PDGFRB fusion genes are responsive to imatinib. SUMMARY: Ongoing research continues to expand our understanding of the pathophysiology of persistent primary hypereosinophilia and clarify the boundaries between some of these disorders. A key challenge is to identify new targets for therapy and limit the number of patients who are classified as having HES.


Subject(s)
Hypereosinophilic Syndrome/diagnosis , Myeloproliferative Disorders/diagnosis , Eosinophilia/metabolism , Humans , Hypereosinophilic Syndrome/metabolism , Myeloproliferative Disorders/metabolism , Protein-Tyrosine Kinases/metabolism
12.
J Leukoc Biol ; 116(2): 297-306, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38682253

ABSTRACT

The noncanonical NF-κB pathway is involved in lymphoid organ development, B-cell maturation, and cytokine production. However, new research has demonstrated that this pathway is also key for the orderly and sequential maturation of myeloid cells, including neutrophils and eosinophils. When this pathway is disrupted or constitutively activated, aberrations in hematopoietic stem and progenitor cell survival and proliferation, as well as subsequent granulopoiesis and eosinophilopoiesis, are affected. Disturbance of such a coordinated and delicate process can manifest in devastating clinical disease, including acute and chronic myeloid leukemias, preleukemic processes such as myelodysplastic syndrome, or hyperinflammatory conditions like hypereosinophilic syndrome. In this review, we discuss the molecular machinery within the noncanonical NF-κB pathway, crosstalk with the canonical NF-κB pathway, murine models of noncanonical signaling, and how aberrations in this pathway manifest in leukemic or hyperinflammatory disease with a focus on hypereosinophilic syndrome. Potential and promising drug therapies will also be discussed, emphasizing the noncanonical NF-κB pathway as a potential target for improved treatment for patients with leukemia or idiopathic hypereosinophilic syndrome. The hope is that review of such mechanisms and treatments may eventually result in findings that aid physicians in rapidly diagnosing and more accurately classifying patients with such complex and overlapping hematopoietic diseases.


Subject(s)
Hypereosinophilic Syndrome , Myelopoiesis , NF-kappa B , Signal Transduction , Humans , NF-kappa B/metabolism , Animals , Hypereosinophilic Syndrome/pathology , Hypereosinophilic Syndrome/metabolism , Lymphocytes/metabolism , Lymphocytes/immunology
13.
Allergy ; 68(6): 713-23, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23621172

ABSTRACT

BACKGROUND: Chronic eosinophilic leukemia (CEL) is a myeloproliferative neoplasm characterized by expansion of neoplastic eosinophils, tissue infiltration, and organ damage. In a subset of these patients, the FIP1L1/PDGFRA (F/P) oncoprotein is detectable. F/P exhibits constitutive tyrosine kinase activity and activates a number of signaling pathways. So far, however, little is known about the role of F/P-dependent proteins in the pathogenesis of CEL. METHODS: A screen for F/P-dependent cytokines was performed in growth factor-dependent human cell lines lentivirally transduced with F/P. Signal transduction pathways were characterized in Ba/F3 cells with doxycycline-inducible expression of F/P and in EOL-1 cells. Cytokine expression was confirmed in patients' material by immunohistochemistry, immunofluorescence, and confocal microscopy. Gene expression analysis, proliferation assays, and chemotaxis assays were used to elucidate paracrine interactions between neoplastic eosinophils and stromal cells. RESULTS: We show that F/P upregulates expression of oncostatin M (OSM) in various cell line models in a STAT5-dependent manner. Correspondingly, neoplastic eosinophils in the bone marrow were found to overexpress OSM. OSM derived from F/P + cells stimulated proliferation of stromal cells. Moreover, OSM-containing supernatants from F/P + cells were found to upregulate production of stromal cell-derived factor-1 (SDF-1)/CXCL12 in human fibroblasts. SDF-1, in turn, induced migration of EOL-1 cells in a dose-dependent manner. CONCLUSIONS: We have identified a F/P-driven paracrine interaction between neoplastic eosinophils and stromal cells that may contribute to tissue fibrosis and accumulation of neoplastic eosinophils in CEL.


Subject(s)
Biomarkers, Tumor/metabolism , Hypereosinophilic Syndrome/metabolism , Oncogene Proteins, Fusion/metabolism , Oncostatin M/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Cell Line , Chemokine CXCL12/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation, Neoplastic , Humans , Hypereosinophilic Syndrome/genetics , Immunoblotting , Immunohistochemistry , Real-Time Polymerase Chain Reaction , STAT5 Transcription Factor/metabolism , Up-Regulation
14.
Allergy ; 67(9): 1149-56, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22775568

ABSTRACT

RATIONALE: Churg-Strauss syndrome (CSS) and hypereosinophilic syndrome (HES) overlap considerably in clinical presentation. A reliable means of distinguishing between these groups of patients is needed, especially in the setting of glucocorticoid therapy. METHODS: A retrospective chart review of 276 adult subjects referred for evaluation of eosinophilia > 1500/µl was performed, and subjects with a documented secondary cause of eosinophilia or a PDGFR -positive myeloproliferative neoplasm were excluded. The remaining subjects were assessed for the presence of American College of Rheumatology (ACR) criteria. Laboratory and clinical parameters were compared between subjects with biopsy-proven vasculitis (CSS; n = 8), ≥4 ACR criteria (probable CSS; n = 21), HES with asthma and/or sinusitis without other CSS-defining criteria (HESwAS; n = 20), HES without asthma or sinusitis (HES; n = 18), and normal controls (n = 8). Serum biomarkers reported to be associated with CSS were measured using standard techniques. RESULTS: There were no differences between the subjects with definite or probable CSS or HES with respect to age, gender, or maintenance steroid dose. Serum CCL17, IL-8, and eotaxin levels were significantly increased in eosinophilic subjects as compared to normal controls, but were similar between the eosinophilic groups. Serum CCL17 correlated with eosinophil count (P < 0.0001, r = 0.73), but not with prednisone dose. CONCLUSIONS: In patients with a history of asthma and sinusitis, distinguishing between ANCA-negative CSS and PDGFR-negative HES is difficult because of significant overlap in clinical presentation and biomarker profiles.


Subject(s)
Biomarkers/blood , Chemokine CCL11/blood , Chemokine CCL17/blood , Churg-Strauss Syndrome/blood , Hypereosinophilic Syndrome/blood , Interleukin-8/blood , Adult , Aged , Aged, 80 and over , Churg-Strauss Syndrome/metabolism , Churg-Strauss Syndrome/pathology , Female , Humans , Hypereosinophilic Syndrome/metabolism , Hypereosinophilic Syndrome/pathology , Male , Middle Aged , Retrospective Studies , Young Adult
15.
J Proteome Res ; 10(4): 1468-80, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21302907

ABSTRACT

The FIP1L1-PDGFRA (F/P) fusion gene, which was identified as a recurrent molecular finding in hypereosinophilic syndrome (HES), lead to a constitutively increased tyrosine kinase activity of the fusion protein. Despite data obtained in animals or cell lines models, the mechanisms underlying the predominant eosinophil lineage targeting and the cytotoxicity of eosinophils in this leukemia remain unclear. To define more precisely intrinsic molecular events associated with F/P gene, we performed a proteomic analysis comparing F/P+ eosinophils (F/P-Eos) and eosinophils from healthy donors (C-Eos). Using 2D-DIGE and mass spectrometry techniques, we identified 41 proteins significantly overexpressed between F/P-Eos and C-Eos. Among them, 17.8% belonged to the oxidoreductase family. We further observed a down-expression of peroxiredoxin-2 (PRX-2) and an overexpression of src-homology-2 domain containing tyrosine phosphatase (SHP-1), enzymes regulating PDGFR downstream pathways, and especially intracellular reactive oxygen species (ROS) production. This profile, confirmed in immunoblot analysis, appears specific to F/P-Eos compared to controls and patients with idiopathic HES. In this clonal disorder possibly involving a pluripotent hematopoietic stem cell, we postulate that the well documented relationships between PDGFRA downstream signals and intracellular ROS levels might influence the phenotype of this leukemia.


Subject(s)
Eosinophils , Hypereosinophilic Syndrome/metabolism , Oncogene Proteins, Fusion/metabolism , Proteome/analysis , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/physiology , mRNA Cleavage and Polyadenylation Factors/metabolism , Adult , Aged , Animals , Cell Line , Databases, Protein , Eosinophils/chemistry , Eosinophils/metabolism , Female , Humans , Hypereosinophilic Syndrome/genetics , Hypereosinophilic Syndrome/physiopathology , Male , Mass Spectrometry/methods , Middle Aged , Oncogene Proteins, Fusion/genetics , Oxidation-Reduction , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Reactive Oxygen Species/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Two-Dimensional Difference Gel Electrophoresis/methods , mRNA Cleavage and Polyadenylation Factors/genetics
16.
Blood ; 114(14): 2969-83, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19608752

ABSTRACT

The clonal CD3(-)CD4(+) T-cell population characterizing lymphocytic variant hypereosinophilic syndrome (L-HES) persists for years, with a subgroup of patients ultimately progressing to T lymphoma. The molecular changes associated with the premalignant clone and the emergence of malignant subclones are unknown, precluding the development of targeted therapy for this HES variant. In this study, we used whole genome arrays to examine gene expression in the CD3(-)CD4(+) T cells and found that 850 genes were differentially regulated during chronic disease compared with CD3(+)CD4(+) T cells from healthy donors. Changes in the expression of 349 genes were altered in association with the clinical progression from chronic L-HES to T lymphoma in 1 patient, with 87 of 349 genes representing further changes in genes whose expression was altered in all chronic disease patients (87 of 850). Array analysis after CD2/CD28-mediated activation revealed that the major gene expression changes observed in the CD3(-)CD4(+) T cells do not reflect activation induced alterations but rather pathways involved in T-cell homeostasis, including transforming growth factor-beta signaling, apoptosis, and T-cell maturation, signaling, and migration. Examination of microRNA expression in the CD3(-)CD4(+) T cells from patients with chronic disease identified 23 microRNAs that changed significantly, among which miR-125a further decreased in association with one patient's evolution to T lymphoma.


Subject(s)
CD3 Complex/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Hypereosinophilic Syndrome/genetics , Hypereosinophilic Syndrome/pathology , Lymphocytes/metabolism , Adolescent , Adult , Female , Flow Cytometry , Gene Expression Profiling , Humans , Hypereosinophilic Syndrome/metabolism , Male , MicroRNAs/physiology , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Young Adult
18.
Cytometry B Clin Cytom ; 100(3): 352-360, 2021 05.
Article in English | MEDLINE | ID: mdl-32157815

ABSTRACT

BACKGROUND: Lymphocytic variant of hypereosinophilic syndrome (L-HES) is a subtype of HES driven by cytokines produced by clonal T-cells. Due to the rarity of its occurrence and challenges in diagnosis, this subtype of HES is under recognized. METHODS AND RESULTS: We report seven patients with L-HES, diagnosed from a group of 136 patients who were referred to our institution for the work-up of hypereosinophilia. The clinical presentation, symptoms and signs were heterogeneous and uncharacteristic; indistinguishable from idiopathic HES. Flow cytometry immunophenotypic analysis revealed aberrant T-cells in all patients, with a Th2 immunophenotype, CD2 + CD3-CD4 + CD5 + CD7dim+/-CD8- in six of seven (86%) cases. CD10 was partially expressed in one of seven (14%) cases, and clonal TCR gene rearrangement was detected by PCR in five of seven (71%) patients. All patients were treated with corticosteroids and two of seven (29%) patients received anti-IL5 antibody therapy. With a median follow-up time of 7.5 years (2.3-14.1 years), one (11%) patient developed peripheral T-cell lymphoma 6.1 years after the initial diagnosis of L-HES and responded well to chemotherapy. All patients were alive at the last follow-up. CONCLUSION: In conclusion, a combination of flow cytometry immunophenotyping and molecular analysis allows the identification of aberrant T-cells, facilitating a diagnosis of L-HES in patients with eosinophilia. A correct diagnosis is essential for the proper management of these patients.


Subject(s)
Hypereosinophilic Syndrome/diagnosis , Hypereosinophilic Syndrome/pathology , T-Lymphocytes/pathology , Adult , Aged , Cytokines/metabolism , Female , Flow Cytometry/methods , Humans , Hypereosinophilic Syndrome/metabolism , Immunophenotyping/methods , Male , Middle Aged , T-Lymphocytes/metabolism
19.
J Allergy Clin Immunol ; 124(6): 1319-25.e3, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19910029

ABSTRACT

BACKGROUND: Hypereosinophilic syndrome (HES) is a heterogeneous group of rare disorders defined by persistent blood eosinophilia > or =1.5 x 10(9)/L, absence of a secondary cause, and evidence of eosinophil-associated pathology. With the exception of a recent multicenter trial of mepolizumab (anti-IL-5 mAb), published therapeutic experience has been restricted to case reports and small case series. OBJECTIVE: The purpose of the study was to collect and summarize baseline demographic, clinical, and laboratory characteristics in a large, diverse cohort of patients with HES and to review responses to treatment with conventional and novel therapies. METHODS: Clinical and laboratory data from 188 patients with HES, seen between January 2001 and December 2006 at 11 institutions in the United States and Europe, were collected retrospectively by chart review. RESULTS: Eighteen of 161 patients (11%) tested were Fip1-like 1-platelet-derived growth factor receptor alpha (FIP1L1-PDGFRA) mutation-positive, and 29 of 168 patients tested (17%) had a demonstrable aberrant or clonal T-cell population. Corticosteroid monotherapy induced complete or partial responses at 1 month in 85% (120/141) of patients with most remaining on maintenance doses (median, 10 mg prednisone equivalent daily for 2 months to 20 years). Hydroxyurea and IFN-alpha (used in 64 and 46 patients, respectively) were also effective, but their use was limited by toxicity. Imatinib (used in 68 patients) was more effective in patients with the FIP1L1-PDGFRA mutation (88%) than in those without (23%; P < .001). CONCLUSION: This study, the largest clinical analysis of patients with HES to date, not only provides useful information for clinicians but also should stimulate prospective trials to optimize treatment of HES.


Subject(s)
Eosinophils/immunology , Hypereosinophilic Syndrome/drug therapy , Oncogene Proteins, Fusion/immunology , Receptor, Platelet-Derived Growth Factor alpha/immunology , mRNA Cleavage and Polyadenylation Factors/immunology , Adolescent , Adrenal Cortex Hormones/administration & dosage , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Benzamides , Chemokine CCL17/blood , Child , Cyclosporine/administration & dosage , Cyclosporine/therapeutic use , Drug Therapy, Combination , Eosinophils/drug effects , Eosinophils/metabolism , Female , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/therapeutic use , Hypereosinophilic Syndrome/immunology , Hypereosinophilic Syndrome/metabolism , Imatinib Mesylate , Interferon-alpha/administration & dosage , Interferon-alpha/therapeutic use , Interleukin-5/blood , Male , Middle Aged , Oncogene Proteins, Fusion/metabolism , Piperazines/administration & dosage , Piperazines/therapeutic use , Pyrimidines/administration & dosage , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Retrospective Studies , Tryptases/blood , Young Adult , mRNA Cleavage and Polyadenylation Factors/metabolism
20.
Clin Rev Allergy Immunol ; 59(2): 231-247, 2020 Oct.
Article in English | MEDLINE | ID: mdl-31919743

ABSTRACT

Eosinophilic inflammation is a component of many atopic diseases such as asthma, and biologics targeting eosinophils have been shown to be effective in subsets of these patients. However, there also are conditions in which eosinophils are the key inflammatory cells responsible for driving tissue damage. In these eosinophilic diseases such as hyper-eosinophilic syndrome, eosinophilic esophagitis, and eosinophilic granulomatosis with polyangiitis (EGPA), the development of biologics inhibiting eosinophilic inflammation have offered targeted therapeutic strategies for patients that have not responded well to typical first line drugs, which often have significant adverse side effects with poor disease modification or recurrent relapse with significant morbidity. IL-5 has long been recognized as the key inflammatory cytokine involved in the priming and survival of eosinophils and their proliferation and maturation in eosinophilic disease. There are a number of trials and case series demonstrating the immunomodulatory benefits of anti-IL-5 therapies in these diseases with good clinical responses. Yet, due to the heterogeneity and rarity of these conditions, anti-IL-5 therapies have not resulted in disease remission for all patients. Clearly, further research into the use of anti-IL-5 therapies in various eosinophilic diseases is needed and ongoing investigation into other immune mechanisms underlying chronic eosinophilic diseases may provide alternative therapies for these challenging conditions.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Eosinophilia/drug therapy , Hypereosinophilic Syndrome/drug therapy , Interleukin-5/antagonists & inhibitors , Molecular Targeted Therapy , Antibodies, Monoclonal/pharmacology , Biomarkers , Clinical Trials as Topic , Diagnosis, Differential , Disease Management , Disease Susceptibility , Eosinophilia/diagnosis , Eosinophilia/etiology , Eosinophilia/metabolism , Humans , Hypereosinophilic Syndrome/diagnosis , Hypereosinophilic Syndrome/etiology , Hypereosinophilic Syndrome/metabolism , Prognosis , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL