Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Health Psychol ; 15(6): 897-904, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20616181

ABSTRACT

This study explored parental experience one year after hematopoietic stem cell transplant for primary immunodeficiency. Eight parents whose child had undergone transplant were interviewed one year after their child's transplant. Transcripts were analysed using interpretative phenomenological analysis. Four themes emerged: parents' paradoxical existence within an 'abnormal normality'; isolation felt by parents; gender differences between mothers and fathers; and the 'positive growth' parents attribute to their experience. As well as describing stressful or traumatic experiences they identified aspects of post-traumatic growth. The methodology used allowed contrasting experiences to emerge and highlights the importance of follow-up for parents as well as children.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immunocompromised Host/immunology , Parents/psychology , Transplantation Conditioning , Child , Child, Preschool , Female , Humans , Interviews as Topic , Male , Parent-Child Relations , United Kingdom
2.
Cancer Cell ; 13(4): 311-20, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18394554

ABSTRACT

We report that TG101348, a selective small-molecule inhibitor of JAK2 with an in vitro IC50 of approximately 3 nM, shows therapeutic efficacy in a murine model of myeloproliferative disease induced by the JAK2V617F mutation. In treated animals, there was a statistically significant reduction in hematocrit and leukocyte count, a dose-dependent reduction/elimination of extramedullary hematopoiesis, and, at least in some instances, evidence for attenuation of myelofibrosis. There were no apparent toxicities and no effect on T cell number. In vivo responses were correlated with surrogate endpoints, including reduction/elimination of JAK2V617F disease burden assessed by quantitative genomic PCR, suppression of endogenous erythroid colony formation, and in vivo inhibition of JAK-STAT signal transduction as assessed by flow cytometric measurement of phosphorylated Stat5.


Subject(s)
Amino Acid Substitution , Disease Models, Animal , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/genetics , Polycythemia Vera/drug therapy , Polycythemia Vera/enzymology , Protein Kinase Inhibitors/therapeutic use , Pyrrolidines/therapeutic use , Sulfonamides/therapeutic use , Animals , Bone Marrow Transplantation , Cell Line, Tumor , Colony-Forming Units Assay , Endpoint Determination , Flow Cytometry , Hematopoietic System/cytology , Hematopoietic System/drug effects , Humans , Janus Kinase 2/metabolism , Mice , Mice, Inbred C57BL , Phenylalanine/genetics , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyrrolidines/pharmacokinetics , Signal Transduction/drug effects , Sulfonamides/pharmacokinetics , Survival Rate , Treatment Outcome , Valine/genetics
3.
Cancer Cell ; 12(4): 367-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17936561

ABSTRACT

Despite their known transforming properties, the effects of leukemogenic FLT3-ITD mutations on hematopoietic stem and multipotent progenitor cells and on hematopoietic differentiation are not well understood. We report a mouse model harboring an ITD in the murine Flt3 locus that develops myeloproliferative disease resembling CMML and further identified FLT3-ITD mutations in a subset of human CMML. These findings correlated with an increase in number, cell cycling, and survival of multipotent stem and progenitor cells in an ITD dose-dependent manner in animals that exhibited alterations within their myeloid progenitor compartments and a block in normal B cell development. This model provides insights into the consequences of constitutive signaling by an oncogenic tyrosine kinase on hematopoietic progenitor quiescence, function, and cell fate.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells/metabolism , Leukemia, Myelomonocytic, Chronic/metabolism , Multipotent Stem Cells/metabolism , Mutation , Myeloproliferative Disorders/metabolism , fms-Like Tyrosine Kinase 3/metabolism , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Exons , Gene Expression Regulation, Neoplastic , Genotype , Hematopoietic Stem Cells/pathology , Humans , Kaplan-Meier Estimate , Leukemia, Experimental/metabolism , Leukemia, Experimental/pathology , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/pathology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Phenotype , Signal Transduction , fms-Like Tyrosine Kinase 3/genetics
4.
Cell ; 128(2): 325-39, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17254970

ABSTRACT

To understand the role of FoxO family members in hematopoiesis, we conditionally deleted FoxO1, FoxO3, and FoxO4 in the adult hematopoietic system. FoxO-deficient mice exhibited myeloid lineage expansion, lymphoid developmental abnormalities, and a marked decrease of the lineage-negative Sca-1+, c-Kit+ (LSK) compartment that contains the short- and long-term hematopoietic stem cell (HSC) populations. FoxO-deficient bone marrow had defective long-term repopulating activity that correlated with increased cell cycling and apoptosis of HSC. Notably, there was a marked context-dependent increase in reactive oxygen species (ROS) in FoxO-deficient HSC compared with wild-type HSC that correlated with changes in expression of genes that regulate ROS. Furthermore, in vivo treatment with the antioxidative agent N-acetyl-L-cysteine resulted in reversion of the FoxO-deficient HSC phenotype. Thus, FoxO proteins play essential roles in the response to physiologic oxidative stress and thereby mediate quiescence and enhanced survival in the HSC compartment, a function that is required for its long-term regenerative potential.


Subject(s)
Blood Cells/metabolism , Cell Differentiation/genetics , Forkhead Transcription Factors/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Oxidative Stress/genetics , Animals , Antioxidants/pharmacology , Blood Cells/cytology , Blood Cells/drug effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/physiopathology , Cell Cycle Proteins , Cell Lineage/drug effects , Cell Lineage/genetics , Cell Survival/drug effects , Cell Survival/genetics , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Gene Expression Regulation/physiology , Hematopoiesis/drug effects , Lymphocytes/cytology , Lymphocytes/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/metabolism , Reactive Oxygen Species/metabolism , Transcription Factors/genetics
5.
Proc Natl Acad Sci U S A ; 103(45): 16924-9, 2006 Nov 07.
Article in English | MEDLINE | ID: mdl-17068127

ABSTRACT

HOX genes have emerged as critical effectors of leukemogenesis, but the mechanisms that regulate their expression in leukemia are not well understood. Recent data suggest that the caudal homeobox transcription factors CDX1, CDX2, and CDX4, developmental regulators of HOX gene expression, may contribute to HOX gene dysregulation in leukemia. We report here that CDX4 is expressed normally in early hematopoietic progenitors and is expressed aberrantly in approximately 25% of acute myeloid leukemia (AML) patient samples. Cdx4 regulates Hox gene expression in the adult murine hematopoietic system and dysregulates Hox genes that are implicated in leukemogenesis. Furthermore, bone marrow progenitors that are retrovirally engineered to express Cdx4 serially replate in methylcellulose cultures, grow in liquid culture, and generate a partially penetrant, long-latency AML in bone marrow transplant recipients. Coexpression of the Hox cofactor Meis1a accelerates the Cdx4 AML phenotype and renders it fully penetrant. Structure-function analysis demonstrates that leukemic transformation requires intact Cdx4 transactivation and DNA-binding domains but not the putative Pbx cofactor interaction motif. Together, these data indicate that Cdx4 regulates Hox gene expression in adult hematopoiesis and may serve as an upstream regulator of Hox gene expression in the induction of acute leukemia. Inasmuch as many human leukemias show dysregulated expression of a spectrum of HOX family members, these collective findings also suggest a central role for CDX4 expression in the genesis of acute leukemia.


Subject(s)
Genes, Homeobox , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/etiology , Neoplasm Proteins/metabolism , Adult , Animals , Base Sequence , Cell Transformation, Neoplastic , Cells, Cultured , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/genetics , Transcriptional Activation
6.
PLoS Med ; 3(7): e270, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16834459

ABSTRACT

BACKGROUND: The JAK2V617F allele has recently been identified in patients with polycythemia vera (PV), essential thrombocytosis (ET), and myelofibrosis with myeloid metaplasia (MF). Subsequent analysis has shown that constitutive activation of the JAK-STAT signal transduction pathway is an important pathogenetic event in these patients, and that enzymatic inhibition of JAK2V617F may be of therapeutic benefit in this context. However, a significant proportion of patients with ET or MF are JAK2V617F-negative. We hypothesized that activation of the JAK-STAT pathway might also occur as a consequence of activating mutations in certain hematopoietic-specific cytokine receptors, including the erythropoietin receptor (EPOR), the thrombopoietin receptor (MPL), or the granulocyte-colony stimulating factor receptor (GCSFR). METHODS AND FINDINGS: DNA sequence analysis of the exons encoding the transmembrane and juxtamembrane domains of EPOR, MPL, and GCSFR, and comparison with germline DNA derived from buccal swabs, identified a somatic activating mutation in the transmembrane domain of MPL (W515L) in 9% (4/45) of JAKV617F-negative MF. Expression of MPLW515L in 32D, UT7, or Ba/F3 cells conferred cytokine-independent growth and thrombopoietin hypersensitivity, and resulted in constitutive phosphorylation of JAK2, STAT3, STAT5, AKT, and ERK. Furthermore, a small molecule JAK kinase inhibitor inhibited MPLW515L-mediated proliferation and JAK-STAT signaling in vitro. In a murine bone marrow transplant assay, expression of MPLW515L, but not wild-type MPL, resulted in a fully penetrant myeloproliferative disorder characterized by marked thrombocytosis (Plt count 1.9-4.0 x 10(12)/L), marked splenomegaly due to extramedullary hematopoiesis, and increased reticulin fibrosis. CONCLUSIONS: Activation of JAK-STAT signaling via MPLW515L is an important pathogenetic event in patients with JAK2V617F-negative MF. The bone marrow transplant model of MPLW515L-mediated myeloproliferative disorders (MPD) exhibits certain features of human MF, including extramedullary hematopoiesis, splenomegaly, and megakaryocytic proliferation. Further analysis of positive and negative regulators of the JAK-STAT pathway is warranted in JAK2V617F-negative MPD.


Subject(s)
Amino Acid Substitution , Mutation, Missense , Point Mutation , Primary Myelofibrosis/genetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Cell Division/drug effects , Cells, Cultured/drug effects , Cells, Cultured/pathology , Colony-Forming Units Assay , Cytokines/pharmacology , Disease Models, Animal , Gene Expression Regulation , Genetic Vectors , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/physiopathology , Hematopoiesis/genetics , Hematopoiesis/physiology , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/physiology , Megakaryocytes/drug effects , Megakaryocytes/pathology , Mice , Mice, Inbred BALB C , Myeloid Cells/drug effects , Myeloid Cells/pathology , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Phosphorylation/drug effects , Primary Myelofibrosis/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/physiology , Receptors, Cytokine/physiology , Recombinant Fusion Proteins/adverse effects , STAT Transcription Factors/physiology , Sequence Analysis, DNA , Signal Transduction/genetics , Signal Transduction/physiology , Spleen/pathology , Thrombocytosis/etiology , Thrombocytosis/genetics , Thrombocytosis/pathology , Transcription, Genetic , mRNA Cleavage and Polyadenylation Factors/genetics
7.
Blood ; 106(9): 3206-13, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16030188

ABSTRACT

AMN107 is a small molecule tyrosine kinase inhibitor developed, in the first instance, as a potent inhibitor of breakpoint cluster region-abelson (BCR-ABL). We tested its effectiveness against fusion tyrosine kinases TEL-platelet-derived growth factor receptorbeta (TEL-PDGFRbeta) and FIP1-like-1 (FIP1L1)-PDGFRalpha, which cause chronic myelomonocytic leukemia and hypereosinophilic syndrome, respectively. In vitro, AMN107 inhibited proliferation of Ba/F3 cells transformed by both TEL-PDGFRbeta and FIP1L1-PDGFRalpha with IC50 (inhibitory concentration 50%) values less than 25 nM and inhibited phosphorylation of the fusion kinases and their downstream signaling targets. The imatinib mesylate-resistant mutant TEL-PDGFRbeta T681I was sensitive to AMN107, whereas the analogous mutation in FIP1L1-PDGFRalpha, T674I, was resistant. In an in vivo bone marrow transplantation assay, AMN107 effectively treated myeloproliferative disease induced by TEL-PDGFRbeta and FIP1L1-PDGFRalpha, significantly increasing survival and disease latency and reducing disease severity as assessed by histopathology and flow cytometry. In summary, AMN107 can inhibit myeloid proliferation driven by TEL-PDGFRbeta and FIP1L1-PDGFRalpha and may be a useful drug for treatment of patients with myeloproliferative disease who harbor these kinase fusions.


Subject(s)
Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor alpha/metabolism , mRNA Cleavage and Polyadenylation Factors/antagonists & inhibitors , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Bone Marrow Transplantation , Cell Line , Disease Models, Animal , Humans , Mice , Mutation/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Oncogene Proteins, Fusion/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Survival Rate , mRNA Cleavage and Polyadenylation Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...