Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 323
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(3)2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38338695

ABSTRACT

Polycythemia vera (PV) and essential thrombocythemia (ET) are myeloproliferative neoplasms (MPN) characterized by clonal erythrocytosis and thrombocytosis, respectively. The main goal of therapy in PV and ET is to prevent thrombohemorrhagic complications. Despite a debated notion that red blood cells (RBCs) play a passive and minor role in thrombosis, there has been increasing evidence over the past decades that RBCs may play a biological and clinical role in PV and ET pathophysiology. This review summarizes the main mechanisms that suggest the involvement of PV and ET RBCs in thrombosis, including quantitative and qualitative RBC abnormalities reported in these pathologies. Among these abnormalities, we discuss increased RBC counts and hematocrit, that modulate blood rheology by increasing viscosity, as well as qualitative changes, such as deformability, aggregation, expression of adhesion proteins and phosphatidylserine and release of extracellular microvesicles. While the direct relationship between a high red cell count and thrombosis is well-known, the intrinsic defects of RBCs from PV and ET patients are new contributors that need to be investigated in depth in order to elucidate their role and pave the way for new therapeutical strategies.


Subject(s)
Polycythemia Vera , Thrombocythemia, Essential , Thrombocytosis , Thrombosis , Humans , Thrombocythemia, Essential/complications , Thrombosis/complications , Thrombocytosis/pathology , Erythrocytes/pathology
2.
J Natl Compr Canc Netw ; 21(12): 1218-1223, 2023 12.
Article in English | MEDLINE | ID: mdl-38081125

ABSTRACT

A 66-year-old male presented with hypereosinophilia, thrombocytosis, extensive thrombosis refractory to direct oral anticoagulant therapy, and evidence of end-organ damage, including rash, splenic infarcts, and pulmonary infiltrates. Bone marrow biopsy revealed myeloid malignancy consistent with both chronic eosinophilic leukemia and myelodysplastic/myeloproliferative neoplasms (MDS/MPN) with SF3B1 mutation and thrombocytosis. Next-generation sequencing of the patient's eosinophils and neutrophil compartments revealed pathologic variants in EZH2 and SF3B1 in addition to a noncanonical JAK2 R683S mutation that has not been previously described in myeloproliferative disorders or other chronic myeloid neoplasms. These mutations were not present in the patient's lymphoid cell fraction, suggesting that the hematopoietic malignancy arose in a myeloid-committed progenitor cell. Based on this case and previous work from our group, we propose that noncanonical JAK2 mutations may permit signal transduction that biases toward eosinophilic differentiation in chronic myeloid neoplasms. Although the patient's blood counts initially responded to ruxolitinib and hydroxyurea, the response was not durable. Early referral for allogenic bone marrow transplant appears necessary to prevent long-term complications and disease progression in myeloid neoplasms with clonal hypereosinophilia driven by noncanonical JAK2 mutations.


Subject(s)
Eosinophilia , Leukemia , Myelodysplastic Syndromes , Myeloproliferative Disorders , Thrombocytosis , Male , Humans , Aged , Diagnosis, Dual (Psychiatry) , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy , Thrombocytosis/diagnosis , Thrombocytosis/genetics , Thrombocytosis/pathology , Mutation , Janus Kinase 2/genetics
3.
Platelets ; 33(8): 1220-1227, 2022 Nov 17.
Article in English | MEDLINE | ID: mdl-35791502

ABSTRACT

The P106L mutation in the human myeloproliferative leukemia virus oncogene (MPL) was shown to be associated with hereditary thrombocythemia in Arabs. The clinical and bone marrow (BM) features of P106L mutation are unknown. Genetic databases at two tertiary hospitals in Saudi Arabia were searched to identify patients with the MPL P106L mutation. Clinical data were collected retrospectively and the BM aspirates and biopsies were independently reviewed by two hematopathologists. In total, 115 patients were included. Median age was 33 years of which 31 patients were pediatric and 65 were female. The mutation was homozygous in 87 patients. Thrombocytosis was documented in 107 patients, with a median platelet count of 667 × 109/L. The homozygous genotype was associated with a higher platelet count. Thirty-three patients had an evaluable BM and clustering of megakaryocytes was observed in 30/33 patients. At the time of last follow-up, 114 patients were alive. The median follow-up was 7.8 years from the time of thrombocytosis. No patients developed disease progression to myelofibrosis. The P106L mutation was associated with marked thrombocytosis at a younger age and with a low risk of thrombosis, splenomegaly, and marrow fibrosis. The BM demonstrated normal or hypocellular marrow with megakaryocyte clusters.


Subject(s)
Primary Myelofibrosis , Receptors, Thrombopoietin , Thrombocytosis , Thrombosis , Adult , Bone Marrow/pathology , Child , Female , Humans , Male , Mutation , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Receptors, Thrombopoietin/genetics , Retrospective Studies , Splenomegaly/genetics , Thrombocytosis/genetics , Thrombocytosis/pathology , Thrombosis/complications
4.
Blood Cells Mol Dis ; 87: 102522, 2021 03.
Article in English | MEDLINE | ID: mdl-33260083

ABSTRACT

Stathmin 1 (STMN1) is a cytosolic phosphoprotein that was discovered as a result of its high level of expression in leukemic cells. It plays an important role in the regulation of mitosis by promoting depolymerization of the microtubules that make up the mitotic spindle and, aging has been shown to impair STMN1 levels and change microtubule stability. We have previously demonstrated that a high level of STMN1 expression during early megakaryopoiesis is necessary for proliferation of megakaryocyte progenitors and that down-regulation of STMN1 expression during late megakaryopoiesis is important for megakaryocyte maturation and platelet production. In this report, we examined the effects of STMN1 deficiency on erythroid and megakaryocytic lineages in the mouse. Our studies show that STMN1 deficiency results in mild thrombocytopenia in young animals which converts into profound thrombocythemia as the mice age. STMN1 deficiency also lead to macrocytic changes in both erythrocytes and megakaryocytes that persisted throughout the life of STMN1 knock-out mice. Furthermore, STMN1 knock-out mice displayed a lower number of erythroid and megakaryocytic progenitor cells and had delayed recovery of their blood counts after chemotherapy. These studies show an important role for STMN1 in normal erythro-megakaryopoietic development and suggests potential implications for disorders affecting these hematopoietic lineages.


Subject(s)
Anemia, Macrocytic/genetics , Erythroid Precursor Cells/pathology , Megakaryocytes/pathology , Stathmin/genetics , Thrombocytosis/genetics , Anemia, Macrocytic/pathology , Animals , Blood Platelets/pathology , Erythropoiesis , Female , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thrombocytosis/pathology
5.
6.
Pediatr Res ; 90(2): 341-346, 2021 08.
Article in English | MEDLINE | ID: mdl-33469189

ABSTRACT

BACKGROUND: Iron deficiency anemia (IDA)-induced reactive thrombocytosis can occur in children and adults. The underlying mechanism for this phenomenon is indeterminate. Traditional cytokines such as thrombopoietin (TPO), interleukin-6 (IL-6), and IL-11 involved in megakaryopoiesis have not been shown to be the cause. Recent studies suggest that growth factors and signaling molecules involved with angiogenesis influence the proliferation and differentiation of megakaryocytes. METHODS: We investigated the possible association between angiogenic cytokines with reactive thrombocytosis due to IDA in an iron-deficient (ID) rat model. Complete blood count, iron panels, and TPO levels were measured at baseline and 5 weeks later in both control (C) and ID rats. Angiogenic cytokines were evaluated in the bone marrow in all rats. RESULTS: We successfully induced IDA in our rats by phlebotomy and reduced iron diet. We did not find an increase of TPO in ID rats. A review of the bone marrow showed an increase in the number of megakaryocytes, vascular structures, as well as increased intensity of stain for vascular endothelial growth factor (VEGF), and CXC chemokine receptor 4 (CXCR4) in rats with IDA compared to controls. CONCLUSIONS: Our results of histological bone marrow data suggest an important role for angiogenesis in the development of IDA-induced thrombocytosis. IMPACT: Thrombocytosis is common with IDA in both children and adults, but the mechanism is unclear. We confirmed that TPO is not the major driver of iron deficiency-associated thrombocytosis. We confirmed the increase in the number of megakaryocytes in the bone marrow despite stable TPO levels. We provided evidence supporting an important role of angiogenesis in megakaryocytopoiesis/thrombopoiesis with increased vascular structures and angiogenic cytokines in the bone marrow of iron-deficient rats. The demonstration that angiogenesis may play an important role in secondary thrombocytosis could lead to a new approach in treating symptomatic reactive thrombocytosis by targeting angiogenesis.


Subject(s)
Anemia, Iron-Deficiency/complications , Bone Marrow/blood supply , Megakaryocytes/metabolism , Neovascularization, Pathologic , Receptors, CXCR4/metabolism , Thrombocytosis/etiology , Thrombopoiesis , Vascular Endothelial Growth Factor A/metabolism , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/pathology , Animals , Disease Models, Animal , Male , Megakaryocytes/pathology , Rats, Sprague-Dawley , Signal Transduction , Thrombocytosis/blood , Thrombocytosis/pathology , Thrombopoietin/metabolism
7.
Platelets ; 32(3): 429-431, 2021 Apr 03.
Article in English | MEDLINE | ID: mdl-32319342

ABSTRACT

Mutations in the calcium channel gene Transient Receptor Potential cation channel subfamily V member 4 (TRPV4) cause autosomal dominant skeletal dysplasia, with phenotypes ranging from mild to perinatal lethality. A recent report detailed enhanced proplatelet formation and increased murine platelet count in the context of TRPV4 activation. No prior reports have described platelet count abnormalities in human TRPV4 disease. Here, we report a case of prolonged thrombocytosis in the context of TRPV4-associated metatropic dysplasia that was lethal in the infantile period.


Subject(s)
TRPV Cation Channels/metabolism , Thrombocytosis/genetics , Female , Humans , Infant , Thrombocytosis/pathology
8.
Genes Chromosomes Cancer ; 59(11): 661-666, 2020 11.
Article in English | MEDLINE | ID: mdl-32557935

ABSTRACT

No specific translocation is associated with myeloproliferative neoplasms (MPNs). However, an interstitial deletion involving subband 17q11.2 which includes the NF1 gene, although rare, is a recurrent aberration in several myeloid disorders including MPNs. For the first time, we report an acquired novel translocation involving 10p13 and 17q11.2 in a 62-year-old Caucasian female which was referred for investigation of chronic and persistent unexplained thrombocytosis. The patient had no history of hematological sequelae and genomic testing for JAK2, CALR, and MPL mutations were negative. She was subsequently diagnosed with a triple negative essential thrombocythemia. Array-CGH analysis noted that the translocation harbored two cryptic deletions, one of which involved 17q11.2 encompassing the NF1 gene. One of the junction breakpoints involved the SUZ12 gene. Immunohistochemical assessment of the marrow trephine showed increased megakaryocytic expression of the SUZ12 protein, as well as EZH2 and Ki67; biochemical abnormalities suggestive of excess megakaryocytic hyperplasia. This novel translocation may affect the expression of SUZ12 and its downstream targets, and may represent a unique pathogenomic etiology which drives chronic thrombocytosis in essential thrombocythemia.


Subject(s)
Neoplasm Proteins/genetics , Thrombocytosis/genetics , Transcription Factors/genetics , Translocation, Genetic , Chromosome Breakpoints , Chromosomes, Human, Pair 10/genetics , Chromosomes, Human, Pair 17/genetics , Comparative Genomic Hybridization , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Gene Deletion , Genetic Testing , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Middle Aged , Neoplasm Proteins/metabolism , Neurofibromin 1/genetics , Thrombocytosis/pathology , Transcription Factors/metabolism
9.
Cardiology ; 145(3): 148-154, 2020.
Article in English | MEDLINE | ID: mdl-32018251

ABSTRACT

INTRODUCTION: Platelets play a fundamental role in the pathogenesis of acute coronary syndrome (ACS). The platelet count (PC) at hospital admission is easy to obtain, but whether thrombocytopenia or/and thrombocytosis impact long-term mortality (LTM) after ACS is unclear. OBJECTIVE: To evaluate the effect of PC at hospital admission on LTM in patients with ACS. METHODS: This retrospective cohort study included patients with the ICD-10 codes for unstable angina (I.20) and acute myocardial infarction (I.21, I.22). Thrombocytopenia was defined as a blood PC <150 G/L and thrombocytosis as a PC >450 G/L. Additional platelet indices which were tested included plateletcrit (PCT), the mean platelet volume (MPV), the platelet distribution width (PDW), and the platelet larger cell ratio (P-LCR). Data on all-cause death were obtained from the National Health Fund database. RESULTS: The study included 3,162 patients with a median follow-up of 27.2 months (interquartile range 12.5-46.8 months; max 68.7 months). Patients with thrombocytopenia and thrombocytosis yielded a higher maximal analyzed 5-year mortality rate in comparison with normal PC patients (45.8 and 47.7 vs. 24.2%, respectively; p < 0.00001) which was mainly driven by higher deaths at 1-2 years after ACS. The 5-year LTM was also significantly higher in patients with abnormal PCT and MPV levels in comparison with patients with PCT and MPV within the normal range. Other platelet indices (PDW, P-LCR) were not associated with a worse outcome. The Cox proportional hazards model revealed that thrombocytopenia at admission was independently associated with higher LTM after ACS (RR 1.83; 95% CI 1.1-3.0; p = 0.01). CONCLUSIONS: Both thrombocytopenia and thrombocytosis at hospital admission in post-ACS patients are associated with a significant almost two times higher 5-year mortality rate.


Subject(s)
Acute Coronary Syndrome/blood , Acute Coronary Syndrome/mortality , Platelet Count , Thrombocytopenia/blood , Thrombocytosis/blood , Adult , Aged , Aged, 80 and over , Blood Platelets/pathology , Cause of Death , Female , Hospitalization , Humans , Male , Mean Platelet Volume , Middle Aged , Poland/epidemiology , Prognosis , Retrospective Studies , Survival Analysis , Thrombocytopenia/pathology , Thrombocytosis/pathology , Time Factors
10.
Int J Mol Sci ; 21(21)2020 Oct 31.
Article in English | MEDLINE | ID: mdl-33142915

ABSTRACT

Platelets are critical components of a number of physiologic processes, including tissue remodeling after injury, wound healing, and maintenance of vascular integrity. Increasing evidence suggests that platelets may also play important roles in cancer. In ovarian cancer, thrombocytosis, both at the time of initial diagnosis and at recurrence, has been associated with poorer prognosis. This review describes current evidence for associations between thrombocytosis and ovarian cancer prognosis and discusses the clinical relevance of platelet count thresholds and timing of assessment. In addition, we discuss several mechanisms from in vitro, in vivo, and clinical studies that may underlie these associations and recommend potential approaches for novel therapeutic targets for this lethal disease.


Subject(s)
Blood Platelets/pathology , Ovarian Neoplasms/pathology , Thrombocytosis/pathology , Female , Humans , Ovarian Neoplasms/blood , Ovarian Neoplasms/etiology , Prognosis , Thrombocytosis/blood
11.
Bull Exp Biol Med ; 169(6): 795-797, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33098506

ABSTRACT

The effects of TLR4 blocker on blood cell morphology, concentrations proinflammatory cytokines, and functional state of the liver and kidneys were studied in outbred male rats (n=60) after intravenous injection of 20 mg/kg LPS isolated from opportunistic Proteus mirabilis strain ATCC 51393. TLR4 blocker TLR4-IN-C34 was injected intravenously in a dose of 1 mg/kg/day over 3 days. Systemic inflammatory reaction induced by LPS was characterized by elevation of serum TNFα, IL-1ß, IL-6, erythrocyte sedimentation rate, leukocytosis, and thrombocytosis. Increased activity of hepatocyte enzymes (ALT, alkaline phosphatase, and lactate dehydrogenase), retention of nitrogen metabolites (urea and creatinine), elevated content of protein oxidation products, and enhanced protein catabolism were also observed. Administration of TLR4 blocker reduced parameters of inflammatory reaction and prevented the development of hypercatabolic syndrome; endotoxicosis and kidney function indicators approached the normal levels.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Leukocytosis/drug therapy , Lipopolysaccharides/antagonists & inhibitors , Pyrans/pharmacology , Sepsis/drug therapy , Thrombocytosis/drug therapy , Toll-Like Receptor 4/antagonists & inhibitors , Alanine Transaminase/blood , Alkaline Phosphatase/blood , Animals , Animals, Outbred Strains , Creatinine/blood , Disease Models, Animal , Gene Expression Regulation , Injections, Intravenous , Interleukin-1beta/blood , Interleukin-1beta/genetics , Interleukin-6/blood , Interleukin-6/genetics , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , L-Lactate Dehydrogenase/blood , Leukocytosis/blood , Leukocytosis/pathology , Lipopolysaccharides/toxicity , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Proteus mirabilis/chemistry , Rats , Sepsis/blood , Sepsis/pathology , Signal Transduction , Thrombocytosis/blood , Thrombocytosis/pathology , Toll-Like Receptor 4/blood , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Urea/blood
13.
Blood ; 129(6): 729-739, 2017 02 09.
Article in English | MEDLINE | ID: mdl-27879260

ABSTRACT

Bone marrow suppression is an adverse effect associated with many antibiotics, especially when administered for prolonged treatment courses. Recent advances in our understanding of steady-state hematopoiesis have allowed us to explore the effects of antibiotics on hematopoietic progenitors in detail using a murine model. Antibiotic-treated mice exhibited anemia, thrombocytosis, and leukopenia, with pronounced pan-lymphopenia as demonstrated by flow cytometric analysis of peripheral blood. Bone marrow progenitor analysis revealed depletion of hematopoietic stem cells and multipotent progenitors across all subtypes. Granulocytes and B cells were also diminished in the bone marrow, whereas the number of CD8+ T cells increased. Reductions in progenitor activity were not observed when cells were directly incubated with antibiotics, suggesting that these effects are indirect. Hematopoietic changes were associated with a significant contraction of the fecal microbiome and were partially rescued by fecal microbiota transfer. Further, mice raised in germ-free conditions had hematopoietic abnormalities similar to those seen in antibiotic-treated mice, and antibiotic therapy of germ-free mice caused no additional abnormalities. The effects of antibiotics were phenocopied in Stat1-deficient mice, with no additional suppression by antibiotics in these mice. We conclude that microbiome depletion as a result of broad-spectrum antibiotic treatment disrupts basal Stat1 signaling and alters T-cell homeostasis, leading to impaired progenitor maintenance and granulocyte maturation. Methods to preserve the microbiome may reduce the incidence of antibiotic-associated bone marrow suppression.


Subject(s)
Anemia/chemically induced , Anti-Bacterial Agents/adverse effects , Gastrointestinal Microbiome/drug effects , Hematopoiesis/drug effects , Leukopenia/chemically induced , STAT1 Transcription Factor/genetics , Thrombocytosis/chemically induced , Anemia/microbiology , Anemia/pathology , Anemia/therapy , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/physiology , Gene Expression , Germ-Free Life/drug effects , Germ-Free Life/genetics , Granulocytes/drug effects , Granulocytes/metabolism , Granulocytes/pathology , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Leukopenia/microbiology , Leukopenia/pathology , Leukopenia/therapy , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , STAT1 Transcription Factor/deficiency , Signal Transduction , Thrombocytosis/microbiology , Thrombocytosis/pathology , Thrombocytosis/therapy
14.
Acta Haematol ; 142(1): 30-36, 2019.
Article in English | MEDLINE | ID: mdl-30970351

ABSTRACT

Inflammatory bowel disease (IBD) is a group of chronic relapsing inflammatory disorders affecting the large and small intestine, with a rising worldwide incidence and prevalence. Anaemia is the most common extraintestinal manifestation of IBD, correlating with disease activity, and tending to relapse even after successful therapy. Iron deficiency is the most common cause; however, it often manifests in combination with anaemia of inflammation. As such, multiple parameters are used for the diagnosis of iron deficiency anaemia in IBD. Timely recognition and selection of appropriate therapy leads to an improvement in the quality of life and prevention of potential sequelae. Oral iron can be effective under specific circumstances; however, as luminal iron changes microbiota and bacterial metabolism, oral administration should be avoided. Intravenous iron is preferred as it bypasses the sites of inflammation. Nevertheless, the optimization of IBD treatment should occur simultaneously, as this improves both patient condition and response to iron therapy. Herein, we discuss the screening, diagnosis, selection of therapy, and follow-up for iron deficiency anaemia in IBD.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Inflammatory Bowel Diseases/pathology , Iron/administration & dosage , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/pathology , Humans , Inflammatory Bowel Diseases/complications , Lymphocytes/cytology , Lymphocytes/metabolism , Quality of Life , Severity of Illness Index , Thrombocytosis/complications , Thrombocytosis/pathology
15.
Blood ; 128(26): 3146-3158, 2016 12 29.
Article in English | MEDLINE | ID: mdl-28034873

ABSTRACT

The mechanisms behind the hereditary thrombocytosis induced by the thrombopoietin (THPO) receptor MPL P106L mutant remain unknown. A complete trafficking defect to the cell surface has been reported, suggesting either weak constitutive activity or nonconventional THPO-dependent mechanisms. Here, we report that the thrombocytosis phenotype induced by MPL P106L belongs to the paradoxical group, where low MPL levels on platelets and mature megakaryocytes (MKs) lead to high serum THPO levels, whereas weak but not absent MPL cell-surface localization in earlier MK progenitors allows response to THPO by signaling and amplification of the platelet lineage. MK progenitors from patients showed no spontaneous growth and responded to THPO, and MKs expressed MPL on their cell surface at low levels, whereas their platelets did not respond to THPO. Transduction of MPL P106L in CD34+ cells showed that this receptor was more efficiently localized at the cell surface on immature than on mature MKs, explaining a proliferative response to THPO of immature cells and a defect in THPO clearance in mature cells. In a retroviral mouse model performed in Mpl-/- mice, MPL P106L could induce a thrombocytosis phenotype with high circulating THPO levels. Furthermore, we could select THPO-dependent cell lines with more cell-surface MPL P106L localization that was detected by flow cytometry and [125I]-THPO binding. Altogether, these results demonstrate that MPL P106L is a receptor with an incomplete defect in trafficking, which induces a low but not absent localization of the receptor on cell surface and a response to THPO in immature MK cells.


Subject(s)
Cell Membrane/metabolism , Mutation/genetics , Receptors, Thrombopoietin/genetics , Thrombocytosis/genetics , Thrombocytosis/pathology , Animals , Cell Line , Disease Models, Animal , Endoplasmic Reticulum Stress , Family , Female , Humans , Male , Megakaryocytes/metabolism , Mice , Pedigree , Protein Transport , Receptors, Thrombopoietin/metabolism , Retroviridae/metabolism , Transduction, Genetic
16.
Blood ; 127(10): 1317-24, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26608331

ABSTRACT

Frameshift mutations in the calreticulin (CALR) gene are seen in about 30% of essential thrombocythemia and myelofibrosis patients. To address the contribution of the CALR mutants to the pathogenesis of myeloproliferative neoplasms, we engrafted lethally irradiated recipient mice with bone marrow cells transduced with retroviruses expressing these mutants. In contrast to wild-type CALR, CALRdel52 (type I) and, to a lesser extent, CALRins5 (type II) induced thrombocytosis due to a megakaryocyte (MK) hyperplasia. Disease was transplantable into secondary recipients. After 6 months, CALRdel52-, in contrast to rare CALRins5-, transduced mice developed a myelofibrosis associated with a splenomegaly and a marked osteosclerosis. Monitoring of virus-transduced populations indicated that CALRdel52 leads to expansion at earlier stages of hematopoiesis than CALRins5. However, both mutants still specifically amplified the MK lineage and platelet production. Moreover, a mutant deleted of the entire exon 9 (CALRdelex9) did not induce a disease, suggesting that the oncogenic property of CALR mutants was related to the new C-terminus peptide. To understand how the CALR mutants target the MK lineage, we used a cell-line model and demonstrated that the CALR mutants, but not CALRdelex9, specifically activate the thrombopoietin (TPO) receptor (MPL) to induce constitutive activation of Janus kinase 2 and signal transducer and activator of transcription 5/3/1. We confirmed in c-mpl- and tpo-deficient mice that expression of Mpl, but not of Tpo, was essential for the CALR mutants to induce thrombocytosis in vivo, although Tpo contributes to disease penetrance. Thus, CALR mutants are sufficient to induce thrombocytosis through MPL activation.


Subject(s)
Calreticulin/metabolism , INDEL Mutation , Megakaryocytes/metabolism , Primary Myelofibrosis/metabolism , Receptors, Thrombopoietin/metabolism , Thrombocytosis/metabolism , Animals , Calreticulin/genetics , Frameshift Mutation , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Megakaryocytes/pathology , Mice , Mice, Mutant Strains , Primary Myelofibrosis/etiology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Receptors, Thrombopoietin/genetics , Thrombocytosis/complications , Thrombocytosis/genetics , Thrombocytosis/pathology
17.
Breast Cancer Res Treat ; 166(3): 819-832, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28831670

ABSTRACT

PURPOSE: Platelets are essential components of hemostasis and also play an important role in the tumor microenvironment. The purposes of our research were to examine the role of thrombocytosis in inflammatory breast cancer (IBC) and to know which cytokine drives thrombocytosis. METHODS: We reviewed the medical records of 3654 patients with stage I-III breast cancer treated between 1998 and 2013, including 230 patients (6%) with IBC. We used Chi-squared test or Fisher's exact test to compare the variables between patients with and without thrombocytosis. Multivariate Cox regression models were used to determine the association of thrombocytosis with overall survival. We also examined baseline serum cytokine levels in 81 patients with primary IBC to determine the association of inflammatory cytokines with thrombocytosis. RESULTS: We found that thrombocytosis was the only variable that predicted prognosis. Fifty-five patients (1.5%) had thrombocytosis. Thrombocytosis was more prevalent in patients with IBC than in those with non-IBC (3.4% vs. 1.4%, p = 0.015). In patients with IBC, thrombocytosis was associated with worse overall survival [hazard ratio 2.38, 95% confidence interval (CI) 1.05-5.4, p = 0.0378]. Circulating levels of growth-regulated oncogene (GRO) (odds ratio 1.003, 95% CI 1.001-1.005, p = 0.0019) and transforming growth factor ß (TGF-ß) (odds ratio 1.3, 95% CI 1.128-1.499, p = 0.0003) were associated with thrombocytosis. CONCLUSIONS: Thrombocytosis was more prevalent in patients with IBC than in those with non-IBC and it was associated with poor prognosis. GRO and TGF-ß were associated with thrombocytosis in IBC.


Subject(s)
Chemokine CXCL1/blood , Inflammatory Breast Neoplasms/blood , Thrombocytosis/blood , Transforming Growth Factor beta1/blood , Adult , Aged , Aged, 80 and over , Blood Platelets/pathology , Cytokines/blood , Female , Humans , Inflammatory Breast Neoplasms/complications , Inflammatory Breast Neoplasms/epidemiology , Inflammatory Breast Neoplasms/pathology , Middle Aged , Neoplasm Staging , Prognosis , Proportional Hazards Models , Thrombocytosis/complications , Thrombocytosis/epidemiology , Thrombocytosis/pathology , Tumor Microenvironment/genetics
19.
Am J Hematol ; 92(1): 94-108, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27991718

ABSTRACT

DISEASE OVERVIEW: Polycythemia Vera (PV) and essential thrombocythemia (ET) are myeloproliferative neoplasms respectively characterized by erythrocytosis and thrombocytosis; other disease features include leukocytosis, splenomegaly, thrombosis, bleeding, microcirculatory symptoms, pruritus, and risk of leukemic or fibrotic transformation. DIAGNOSIS: PV is defined by a JAK2 mutation, whose absence, combined with normal or increased serum erythropoietin level, makes the diagnosis unlikely. JAK2, CALR, and MPL mutations are the mutually exclusive "driver" mutations in ET with respective incidences of 55%, 25%, and 3%; approximately 17% are triple-negative. However, the same molecular markers might also be present in prefibrotic myelofibrosis, whose morphological distinction from ET is prognostically relevant. SURVIVAL AND LEUKEMIC/FIBROTIC TRANSFORMATION: Median survivals are approximately 14 years for PV and 20 years for ET; the corresponding values for younger patients (age <60 years) are 24 and 33 years. Life-expectancy in ET is inferior to the control population. Driver mutational status has not been shown to affect survival in ET whereas the presence of JAK2/MPL mutations has been associated with higher risk of arterial thrombosis and that of MPL with higher risk of fibrotic progression. Risk factors for overall survival in both ET and PV include advanced age, leukocytosis and thrombosis. Leukemic transformation rates at 20 years are estimated at <10% for PV and 5% for ET; fibrotic transformation rates are slightly higher. Most recently, ASXL1, SRSF2, and IDH2 mutations have been associated with inferior overall, leukemia-free or fibrosis-free survival in PV; similarly adverse mutations in ET included SH2B3, SF3B1, U2AF1, TP53, IDH2, and EZH2. THROMBOSIS RISK STRATIFICATION: Current risk stratification in PV and ET is designed to estimate the likelihood of recurrent thrombosis. Accordingly, PV includes two risk categories: high-risk (age >60 years or thrombosis history) and low-risk (absence of both risk factors). In ET, risk stratification includes four categories: very low risk (age ≤60 years, no thrombosis history, JAK2/MPL un-mutated), low risk (age ≤60 years, no thrombosis history, JAK2/MPL mutated), intermediate risk (age >60 years, no thrombosis history, JAK2/MPL un-mutated), and high risk (thrombosis history or age >60 years with JAK2/MPL mutation). In addition, presence of extreme thrombocytosis (platelets >1000 × 10(9)/L) might be associated with acquired von Willebrand syndrome (AvWS) and, therefore, risk of bleeding. RISK-ADAPTED THERAPY: The main goal of therapy in PV and ET is to prevent thrombohemorrhagic complications. All patients with PV require phlebotomy to keep hematocrit below 45% and once-daily aspirin (81 mg). In addition, high-risk patients with PV require cytoreductive therapy. Very low risk ET patients might not require any form of therapy while low-risk patients require at least once-daily aspirin therapy. Cytoreductive therapy is also recommended for high-risk ET patients but it is not mandatory for intermediate-risk patients. First-line drug of choice for cytoreductive therapy, in both ET and PV, is hydroxyurea and second-line drugs of choice are interferon-α and busulfan. We currently do not recommend treatment with ruxolutinib or other JAK2 inhibitors in PV or ET, unless in the presence of severe and protracted pruritus or marked splenomegaly that is not responding to the aforementioned drugs. Screening for AvWS is recommended before administrating aspirin, in the presence of extreme thrombocytosis. Am. J. Hematol. 92:95-108, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Polycythemia Vera/diagnosis , Polycythemia Vera/drug therapy , Thrombocythemia, Essential/diagnosis , Thrombocythemia, Essential/drug therapy , Aspirin/administration & dosage , Aspirin/therapeutic use , Calreticulin/genetics , Diagnosis, Differential , Erythropoietin/blood , Humans , Hydroxyurea/administration & dosage , Hydroxyurea/therapeutic use , Interferon-alpha/administration & dosage , Interferon-alpha/therapeutic use , Janus Kinase 2/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mutation , Polycythemia Vera/blood , Polycythemia Vera/genetics , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/mortality , Primary Myelofibrosis/pathology , Receptors, Thrombopoietin/genetics , Risk Assessment , Thrombocythemia, Essential/blood , Thrombocythemia, Essential/genetics , Thrombocytosis/diagnosis , Thrombocytosis/mortality , Thrombocytosis/pathology
20.
J Immunol ; 194(2): 750-60, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25505285

ABSTRACT

IL-33 is a cytokine of the IL-1 family, which signals through the ST2 receptor. Previous studies emphasized a role for IL-33 in shaping innate and adaptive immune responses. IL-33 was also reported to modulate myelopoiesis and myeloid cell activity. In this article, we describe IL-33-overexpressing CMV/IL33 and LysM/IL33 mice, which display an inflammatory phenotype associated with growth retardation and paw swelling. The phenotype of CMV/IL33 mice is dependent on activation of the ST2 receptor and is characterized by extensive neutrophil infiltration into different organs, including the paws. Local or systemic levels of proinflammatory mediators such as IL-1ß, Cxcl-1, G-CSF, and IL-6 are increased. CMV/IL-33 mice also suffer from anemia, thrombocytosis, and a marked dysregulation of myelopoiesis, leading to an important increase in myeloid cell production or accumulation in bone marrow (BM), spleen, and peripheral blood. Consistently, recombinant IL-33 induced proliferation of myeloid lineage cells in BM-derived granulocyte cultures, whereas IL-33 knockout mice exhibited minor deficiencies in spleen and BM myeloid cell populations. Our observations reveal a neutrophil-dominated inflammatory phenotype in IL-33-overexpressing CMV/IL33 and LysM/IL33 mice, and highlight important regulatory effects of IL-33 on myelopoiesis in vitro and in vivo, where excessive IL-33 signaling can translate into the occurrence of a myeloproliferative disorder.


Subject(s)
Interleukins/immunology , Myelopoiesis/immunology , Neutrophil Infiltration/immunology , Neutrophils/immunology , Signal Transduction/immunology , Anemia/genetics , Anemia/immunology , Anemia/pathology , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Gene Expression , Granulocyte Colony-Stimulating Factor/genetics , Granulocyte Colony-Stimulating Factor/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-1 Receptor-Like 1 Protein , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-33 , Interleukin-6/genetics , Interleukin-6/immunology , Interleukins/genetics , Mice , Mice, Knockout , Myelopoiesis/genetics , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/immunology , Myeloproliferative Disorders/pathology , Neutrophil Infiltration/genetics , Neutrophils/pathology , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Signal Transduction/genetics , Thrombocytosis/genetics , Thrombocytosis/immunology , Thrombocytosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL