Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Mol Ther ; 30(7): 2505-2521, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35443935

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is an aggressive myeloproliferative neoplasia that lacks effective targeted chemotherapies. Clinically, JMML manifests as monocytic leukocytosis, splenomegaly with consequential thrombocytopenia. Most commonly, patients have gain-of-function (GOF) oncogenic mutations in PTPN11 (SHP2), leading to Erk and Akt hyperactivation. Mechanism(s) involved in co-regulation of Erk and Akt in the context of GOF SHP2 are poorly understood. Here, we show that Bruton's tyrosine kinase (BTK) is hyperphosphorylated in GOF Shp2-bearing cells and utilizes B cell adaptor for PI3K to cooperate with p110δ, the catalytic subunit of PI3K. Dual inhibition of BTK and p110δ reduces the activation of both Erk and Akt. In vivo, individual targeting of BTK or p110δ in a mouse model of human JMML equally reduces monocytosis and splenomegaly; however, the combined treatment results in a more robust inhibition and uniquely rescues anemia and thrombocytopenia. RNA-seq analysis of drug-treated mice showed a profound reduction in the expression of genes associated with leukemic cell migration and inflammation, leading to correction in the infiltration of leukemic cells in the lung, liver, and spleen. Remarkably, in a patient derived xenograft model of JMML, leukemia-initiating stem and progenitor cells were potently inhibited in response to the dual drug treatment.


Subject(s)
Leukemia, Myelomonocytic, Juvenile , Thrombocytopenia , Agammaglobulinaemia Tyrosine Kinase/genetics , Animals , Humans , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/metabolism , Leukemia, Myelomonocytic, Juvenile/therapy , Mice , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt/metabolism , Splenomegaly/genetics , Stem Cells/metabolism
2.
Br J Haematol ; 192(4): 720-728, 2021 02.
Article in English | MEDLINE | ID: mdl-32599655

ABSTRACT

Comorbidities influence survival in patients with chronic lymphocytic leukaemia (CLL) treated with chemo-immunotherapy or ibrutinib. While idelalisib has been studied in patients with comorbidities, their impact has not been investigated. We analysed 481 patients treated with idelalisib on two randomised trials (NCT01659021 and NCT01539512). Comorbidities were assessed using the Cumulative Illness Risk Scale (CIRS). Patients received idelalisib + anti-CD20 (rituximab or ofatumumab; n = 284) or anti-CD20 alone (n = 197). The median age was 69 years. We found that comorbidities did not significantly affect outcomes of idelalisib therapy. The objective response rate (ORR) was 79·3% versus 85·8%, the median progression-free survival (PFS) was 16·3 versus 19·1 months, and the median overall survival (OS) was 39·8 versus 49·8 months in patients treated with idelalisib who had a CIRS score of >6 versus ≤6, correspondingly. Treatment with idelalisib + anti-CD20 was associated with superior PFS and ORR when compared to anti-CD20 monotherapy in patients who had high comorbidities (CIRS score of >6) or at least one severe comorbidity (median PFS 16·3 vs. 6·9 months and 16·6 vs. 6·5 months; odds ratio 20·1 and 33·2; P < 0·0001). Thus, comorbidities do not portend inferior outcomes in patients with CLL treated with idelalisib in combination with anti-CD20 therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Purines/therapeutic use , Quinazolinones/therapeutic use , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Comorbidity , Disease-Free Survival , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/epidemiology , Male , Middle Aged , Progression-Free Survival , Rituximab/therapeutic use , Treatment Outcome
3.
Dev Dyn ; 246(12): 1001-1014, 2017 12.
Article in English | MEDLINE | ID: mdl-28975680

ABSTRACT

BACKGROUND: Accumulating evidence suggests the origin of juvenile myelomonocytic leukemia (JMML) is closely associated with fetal development. Nevertheless, the contribution of embryonic progenitors to JMML pathogenesis remains unexplored. We hypothesized that expression of JMML-initiating PTPN11 mutations in HSC-independent yolk sac erythromyeloid progenitors (YS EMPs) would result in a mouse model of pediatric myeloproliferative neoplasm (MPN). RESULTS: E9.5 YS EMPs from VavCre+;PTPN11D61Y embryos demonstrated growth hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF) and hyperactive RAS-ERK signaling. Mutant EMPs engrafted the spleens of neonatal recipients, but did not cause disease. To assess MPN development during unperturbed hematopoiesis we generated CSF1R-MCM+;PTPN11E76K ;ROSAYFP mice in which oncogene expression was restricted to EMPs. Yellow fluorescent protein-positive progeny of mutant EMPs persisted in tissues one year after birth and demonstrated hyperactive RAS-ERK signaling. Nevertheless, these mice had normal survival and did not demonstrate features of MPN. CONCLUSIONS: YS EMPs expressing mutant PTPN11 demonstrate functional and molecular features of JMML but do not cause disease following transplantation nor following unperturbed development. Developmental Dynamics 246:1001-1014, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Erythroid Precursor Cells/enzymology , Gain of Function Mutation , Leukemia, Myelomonocytic, Juvenile/enzymology , MAP Kinase Signaling System , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Yolk Sac/metabolism , Animals , Erythroid Precursor Cells/pathology , Erythroid Precursor Cells/transplantation , Leukemia, Myelomonocytic, Juvenile/embryology , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/pathology , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Yolk Sac/pathology
4.
J Biol Chem ; 290(7): 3894-909, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25538234

ABSTRACT

Macrophages are vital to innate immunity and express pattern recognition receptors and integrins for the rapid detection of invading pathogens. Stimulation of Dectin-1 and complement receptor 3 (CR3) activates Erk- and Akt-dependent production of reactive oxygen species (ROS). Shp2, a protein-tyrosine phosphatase encoded by Ptpn11, promotes activation of Ras-Erk and PI3K-Akt and is crucial for hematopoietic cell function; however, no studies have examined Shp2 function in particulate-stimulated ROS production. Maximal Dectin-1-stimulated ROS production corresponded kinetically to maximal Shp2 and Erk phosphorylation. Bone marrow-derived macrophages (BMMs) from mice with a conditionally deleted allele of Ptpn11 (Shp2(flox/flox);Mx1Cre+) produced significantly lower ROS levels compared with control BMMs. Although YFP-tagged phosphatase dead Shp2-C463A was strongly recruited to the early phagosome, its expression inhibited Dectin-1- and CR3-stimulated phospho-Erk and ROS levels, placing Shp2 phosphatase function and Erk activation upstream of ROS production. Further, BMMs expressing gain of function Shp2-D61Y or Shp2-E76K and peritoneal exudate macrophages from Shp2D61Y/+;Mx1Cre+ mice produced significantly elevated levels of Dectin-1- and CR3-stimulated ROS, which was reduced by pharmacologic inhibition of Erk. SIRPα (signal regulatory protein α) is a myeloid inhibitory immunoreceptor that requires tyrosine phosphorylation to exert its inhibitory effect. YFP-Shp2C463A-expressing cells have elevated phospho-SIRPα levels and an increased Shp2-SIRPα interaction compared with YFP-WT Shp2-expressing cells. Collectively, these findings indicate that Shp2 phosphatase function positively regulates Dectin-1- and CR3-stimulated ROS production in macrophages by dephosphorylating and thus mitigating the inhibitory function of SIRPα and by promoting Erk activation.


Subject(s)
Macrophages/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Reactive Oxygen Species/metabolism , Respiratory Burst/physiology , Animals , Blotting, Western , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fluorescent Antibody Technique , Immunoprecipitation , Integrases/metabolism , Lectins, C-Type/metabolism , Macrophages/cytology , Male , Mice , Mice, Knockout , Phagocytosis , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tyrosine/metabolism
5.
Blood ; 123(18): 2838-42, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24553178

ABSTRACT

Although hyperactivation of the Ras-Erk signaling pathway is known to underlie the pathogenesis of juvenile myelomonocytic leukemia (JMML), a fatal childhood disease, the PI3K-Akt signaling pathway is also dysregulated in this disease. Using genetic models, we demonstrate that inactivation of phosphatidylinositol-3-kinase (PI3K) catalytic subunit p110δ, but not PI3K p110α, corrects gain-of-function (GOF) Shp2-induced granulocyte macrophage-colony-stimulating factor (GM-CSF) hypersensitivity, Akt and Erk hyperactivation, and skewed hematopoietic progenitor distribution. Likewise, potent p110δ-specific inhibitors curtail the proliferation of GOF Shp2-expressing hematopoietic cells and cooperate with mitogen-activated or extracellular signal-regulated protein kinase kinase (MEK) inhibition to reduce proliferation further and maximally block Erk and Akt activation. Furthermore, the PI3K p110δ-specific inhibitor, idelalisib, also demonstrates activity against primary leukemia cells from individuals with JMML. These findings suggest that selective inhibition of the PI3K catalytic subunit p110δ could provide an innovative approach for treatment of JMML, with the potential for limiting toxicity resulting from the hematopoietic-restricted expression of p110δ.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Leukemia, Myelomonocytic, Juvenile/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Proliferation/drug effects , Class Ia Phosphatidylinositol 3-Kinase/genetics , Disease Models, Animal , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Leukemia, Myelomonocytic, Juvenile/genetics , Mice , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
6.
Blood ; 121(24): 4815-7, 2013 Jun 13.
Article in English | MEDLINE | ID: mdl-23766458

ABSTRACT

In this issue of Blood, Gandre-Babbe et al have, in part, overcome the obstacle of validating the molecular underpinnings of juvenile myelomonocytic leukemia (JMML) with the generation of induced pluripotent stem cells (iPSCs) from individuals with JMML.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Leukemia, Myelomonocytic, Juvenile/metabolism , Mutation, Missense , Neoplastic Stem Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Female , Humans , Male
7.
Cytotherapy ; 17(5): 579-92, 2015 May.
Article in English | MEDLINE | ID: mdl-25559145

ABSTRACT

BACKGROUND AIMS: Human cord blood (CB) is enriched in circulating endothelial colony forming cells (ECFCs) that display high proliferative potential and in vivo vessel forming ability. Because Notch signaling is critical for embryonic blood vessel formation in utero, we hypothesized that Notch pathway activation may enhance cultured ECFC vasculogenic properties in vivo. METHODS: In vitro ECFC stimulation with an immobilized chimeric Notch ligand (Delta-like1(ext-IgG)) led to significant increases in the mRNA and protein levels of Notch regulated Hey2 and EphrinB2 that were blocked by treatment with γ-secretase inhibitor addition. However, Notch stimulated preconditioning in vitro failed to enhance ECFC vasculogenesis in vivo. In contrast, in vivo co-implantation of ECFCs with OP9-Delta-like 1 stromal cells that constitutively expressed the Notch ligand delta-like 1 resulted in enhanced Notch activated ECFC-derived increased vessel density and enlarged vessel area in vivo, an effect not induced by OP9 control stromal implantation. RESULTS: This Notch activation was associated with diminished apoptosis in the exposed ECFC. CONCLUSIONS: We conclude that Notch pathway activation in ECFC in vivo via co-implanted stromal cells expressing delta-like 1 promotes vasculogenesis and augments blood vessel formation via diminishing apoptosis of the implanted ECFC.


Subject(s)
Endothelial Cells/cytology , Fetal Blood/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Receptors, Notch/metabolism , Animals , Calcium-Binding Proteins , Cells, Cultured , Collagen/pharmacology , Colony-Forming Units Assay , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Infant, Newborn , Ligands , Mice, Inbred NOD , Mice, SCID , Neovascularization, Physiologic/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Stromal Cells/cytology , Stromal Cells/metabolism
8.
Development ; 138(2): 303-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21177343

ABSTRACT

Dishevelled-associated activator of morphogenesis 1 (Daam1), a member of the formin protein family, plays an important role in regulating the actin cytoskeleton via mediation of linear actin assembly. Previous functional studies of Daam1 in lower species suggest its essential role in Drosophila trachea formation and Xenopus gastrulation. However, its in vivo physiological function in mammalian systems is largely unknown. We have generated Daam1-deficient mice via gene-trap technology and found that Daam1 is highly expressed in developing murine organs, including the heart. Daam1-deficient mice exhibit embryonic and neonatal lethality and suffer multiple cardiac defects, including ventricular noncompaction, double outlet right ventricles and ventricular septal defects. In vivo genetic rescue experiments further confirm that the lethality of Daam1-deficient mice results from the inherent cardiac abnormalities. In-depth analyses have revealed that Daam1 is important for regulating filamentous actin assembly and organization, and consequently for cytoskeletal function in cardiomyocytes, which contributes to proper heart morphogenesis. Daam1 is also found to be important for proper cytoskeletal architecture and functionalities in embryonic fibroblasts. Biochemical analyses indicate that Daam1 does not regulate cytoskeletal organization through RhoA, Rac1 or Cdc42. Our study highlights a crucial role for Daam1 in regulating the actin cytoskeleton and tissue morphogenesis.


Subject(s)
Fetal Heart/embryology , Microfilament Proteins/physiology , rho GTP-Binding Proteins/physiology , Actins/metabolism , Animals , Apoptosis , Base Sequence , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cells, Cultured , DNA Primers/genetics , Female , Fetal Heart/abnormalities , Fetal Heart/cytology , Fetal Heart/metabolism , Gene Expression Regulation, Developmental , Heart Defects, Congenital/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/deficiency , Microfilament Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Morphogenesis/genetics , Morphogenesis/physiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phenotype , Pregnancy , rho GTP-Binding Proteins/deficiency , rho GTP-Binding Proteins/genetics
9.
Blood ; 120(13): 2669-78, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22806893

ABSTRACT

Intracellular mechanism(s) that contribute to promiscuous signaling via oncogenic KIT in systemic mastocytosis and acute myelogenous leukemia are poorly understood. We show that SHP2 phosphatase is essential for oncogenic KIT-induced growth and survival in vitro and myeloproliferative disease (MPD) in vivo. Genetic disruption of SHP2 or treatment of oncogene-bearing cells with a novel SHP2 inhibitor alone or in combination with the PI3K inhibitor corrects MPD by disrupting a protein complex involving p85α, SHP2, and Gab2. Importantly, a single tyrosine at position 719 in oncogenic KIT is sufficient to develop MPD by recruiting p85α, SHP2, and Gab2 complex to oncogenic KIT. Our results demonstrate that SHP2 phosphatase is a druggable target that cooperates with lipid kinases in inducing MPD.


Subject(s)
Cell Transformation, Neoplastic/pathology , GRB2 Adaptor Protein/physiology , Mutation/genetics , Myeloproliferative Disorders/etiology , Myeloproliferative Disorders/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Proto-Oncogene Proteins c-kit/genetics , Animals , Apoptosis , Blotting, Western , Bone Marrow Transplantation , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Immunoprecipitation , Integrases/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Myeloproliferative Disorders/mortality , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/metabolism , Signal Transduction/drug effects , Survival Rate , Tyrosine/metabolism
10.
Proc Natl Acad Sci U S A ; 108(46): E1146-55, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22006328

ABSTRACT

Autosomal dominant hypophosphatemic rickets (ADHR) is unique among the disorders involving Fibroblast growth factor 23 (FGF23) because individuals with R176Q/W and R179Q/W mutations in the FGF23 (176)RXXR(179)/S(180) proteolytic cleavage motif can cycle from unaffected status to delayed onset of disease. This onset may occur in physiological states associated with iron deficiency, including puberty and pregnancy. To test the role of iron status in development of the ADHR phenotype, WT and R176Q-Fgf23 knock-in (ADHR) mice were placed on control or low-iron diets. Both the WT and ADHR mice receiving low-iron diet had significantly elevated bone Fgf23 mRNA. WT mice on a low-iron diet maintained normal serum intact Fgf23 and phosphate metabolism, with elevated serum C-terminal Fgf23 fragments. In contrast, the ADHR mice on the low-iron diet had elevated intact and C-terminal Fgf23 with hypophosphatemic osteomalacia. We used in vitro iron chelation to isolate the effects of iron deficiency on Fgf23 expression. We found that iron chelation in vitro resulted in a significant increase in Fgf23 mRNA that was dependent upon Mapk. Thus, unlike other syndromes of elevated FGF23, our findings support the concept that late-onset ADHR is the product of gene-environment interactions whereby the combined presence of an Fgf23-stabilizing mutation and iron deficiency can lead to ADHR.


Subject(s)
Familial Hypophosphatemic Rickets/genetics , Fibroblast Growth Factors/genetics , Iron Deficiencies , Anemia, Iron-Deficiency/complications , Animals , Familial Hypophosphatemic Rickets/physiopathology , Female , Fibroblast Growth Factor-23 , Gene-Environment Interaction , Glucuronidase/metabolism , Hypophosphatemia/genetics , Klotho Proteins , MAP Kinase Signaling System , Male , Mice , Mice, Transgenic , Osteocytes/cytology , Osteomalacia/genetics , Phenotype , Protein Structure, Tertiary , Rats
11.
J Am Soc Nephrol ; 24(1): 113-24, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23222126

ABSTRACT

In the rat, p53 promotes tubular apoptosis after ischemic AKI. Acute pharmacologic inhibition of p53 is protective in this setting, but chronic inhibition enhances fibrosis, demonstrating that the role of p53 in ischemic AKI is incompletely understood. Here, we investigated whether genetic absence of p53 is also protective in ischemic AKI. Surprisingly, p53-knockout mice (p53(-/-)) had worse kidney injury, compared with wild-type mice, and exhibited increased and prolonged infiltration of leukocytes after ischemia. Acute inhibition of p53 with pifithrin-α in wild-type mice mimicked the observations in p53(-/-) mice. Chimeric mice that lacked p53 in leukocytes sustained injury similar to p53(-/-) mice, suggesting an important role for leukocyte p53 in ischemic AKI. Compared with wild-type mice, a smaller proportion of macrophages in the kidneys of p53(-/-) and pifithrin-α-treated mice after ischemic injury were the anti-inflammatory M2 phenotype. Ischemic kidneys of p53(-/-) and pifithrin-α-treated mice also showed reduced expression of Kruppel-like factor-4. Finally, models of peritonitis in p53(-/-) and pifithrin-α-treated mice confirmed the anti-inflammatory role of p53 and its effect on the polarization of macrophage phenotype. In summary, in contrast to the rat, inflammation characterizes ischemic AKI in mice; leukocyte p53 is protective by reducing the extent and duration of this inflammation and by promoting the anti-inflammatory M2 macrophage phenotype.


Subject(s)
Acute Kidney Injury/metabolism , Inflammation/metabolism , Kidney/metabolism , Reperfusion Injury/metabolism , Tumor Suppressor Protein p53/metabolism , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Animals , Apoptosis , Benzothiazoles , Bone Marrow Transplantation , Chimera , Cytokines/metabolism , Disease Models, Animal , Kidney/blood supply , Kidney/pathology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Leukocytes/physiology , Macrophages/pathology , Male , Mice , Peritonitis/metabolism , Phenotype , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Toluene/analogs & derivatives , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics
12.
Blood ; 118(13): 3459-69, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21791431

ABSTRACT

Mast cell maturation is poorly understood. We show that enhanced PI3K activation results in accelerated maturation of mast cells by inducing the expression of microphthalmia transcription factor (Mitf). Conversely, loss of PI3K activation reduces the maturation of mast cells by inhibiting the activation of AKT, leading to reduced Mitf but enhanced Gata-2 expression and accumulation of Gr1(+)Mac1(+) myeloid cells as opposed to mast cells. Consistently, overexpression of Mitf accelerates the maturation of mast cells, whereas Gata-2 overexpression mimics the loss of the PI3K phenotype. Expressing the full-length or the src homology 3- or BCR homology domain-deleted or shorter splice variant of the p85α regulatory subunit of PI3K or activated AKT or Mitf in p85α-deficient cells restores the maturation but not growth. Although deficiency of both SHIP and p85α rescues the maturation of SHIP(-/-) and p85α(-/-) mast cells and expression of Mitf; in vivo, mast cells are rescued in some, but not all tissues, due in part to defective KIT signaling, which is dependent on an intact src homology 3 and BCR homology domain of p85α. Thus, p85α-induced maturation, and growth and survival signals, in mast cells can be uncoupled.


Subject(s)
Cell Differentiation/genetics , Mast Cells/physiology , Microphthalmia-Associated Transcription Factor/physiology , Phosphatidylinositol 3-Kinases/physiology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Cell Differentiation/physiology , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Transfection
13.
Proc Natl Acad Sci U S A ; 107(10): 4573-8, 2010 Mar 09.
Article in English | MEDLINE | ID: mdl-20167798

ABSTRACT

Protein tyrosine phosphatases are often exploited and subverted by pathogenic bacteria to cause human diseases. The tyrosine phosphatase mPTPB from Mycobacterium tuberculosis is an essential virulence factor that is secreted by the bacterium into the cytoplasm of macrophages, where it mediates mycobacterial survival in the host. Consequently, there is considerable interest in understanding the mechanism by which mPTPB evades the host immune responses, and in developing potent and selective mPTPB inhibitors as unique antituberculosis (antiTB) agents. We uncovered that mPTPB subverts the innate immune responses by blocking the ERK1/2 and p38 mediated IL-6 production and promoting host cell survival by activating the Akt pathway. We identified a potent and selective mPTPB inhibitor I-A09 with highly efficacious cellular activity, from a combinatorial library of bidentate benzofuran salicylic acid derivatives assembled by click chemistry. We demonstrated that inhibition of mPTPB with I-A09 in macrophages reverses the altered host immune responses induced by the bacterial phosphatase and prevents TB growth in host cells. The results provide the necessary proof-of-principle data to support the notion that specific inhibitors of the mPTPB may serve as effective antiTB therapeutics.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Macrophages/drug effects , Protein Tyrosine Phosphatases/antagonists & inhibitors , Animals , Antitubercular Agents/chemical synthesis , Antitubercular Agents/chemistry , Apoptosis/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Benzofurans/chemical synthesis , Benzofurans/chemistry , Benzofurans/pharmacology , Cell Line , Cell Survival/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Host-Pathogen Interactions , Humans , Interferon-gamma/metabolism , Kinetics , Macrophages/metabolism , Macrophages/microbiology , Mice , Microbial Sensitivity Tests , Mitogen-Activated Protein Kinases/metabolism , Models, Chemical , Molecular Structure , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/physiology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Triazoles/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacology , Tuberculosis/microbiology , Tuberculosis/prevention & control
14.
Curr Opin Hematol ; 19(4): 273-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22504523

ABSTRACT

PURPOSE OF REVIEW: The protein tyrosine phosphatase Shp2 is encoded by PTPN11 and positively regulates physiologic hematopoiesis. Mutations of PTPN11 cause the congenital disorder Noonan syndrome and pathologically promote human leukemias. Given the high frequency of PTPN11 mutations in human disease, several animal models have been generated to investigate Shp2 in hematopoietic stem cell (HSC) function and leukemic transformation. RECENT FINDINGS: Two independent animal models bearing knockout of Shp2 in hematopoietic tissues clearly demonstrate the necessity of Shp2 in HSC repopulating capacity. Reduced HSC quiescence and increased apoptosis accounts for diminished HSC function in the absence of Shp2. The germline mutation Shp2D61G enhances HSC activity and induces myeloproliferative disease (MPD) in vivo by HSC transformation. The somatic mutation Shp2D61Y produces MPD in vivo but fails to induce acute leukemia, whereas somatic Shp2E76K produces MPD in vivo that transforms into full-blown leukemia. HSCs expressing Shp2D61Y do not generate MPD in recipient animals upon transplantation, whereas Shp2E76K-expressing HSCs yield MPD as well as acute leukemia in recipient animals. The mechanisms underlying the unique functions of Shp2D61Y and Shp2E76K in HSC transformation and leukemogenesis continue to be under investigation. SUMMARY: Further understanding of the physiologic and pathologic role of Shp2 in hematopoiesis and leukemogenesis, respectively, will yield information needed to develop therapeutic strategies targeted to Shp2 in human disease.


Subject(s)
Cell Transformation, Neoplastic , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/enzymology , Myeloproliferative Disorders/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/physiology , Animals , Gene Expression Regulation, Neoplastic/genetics , Hematopoietic Stem Cells/cytology , Leukemia, Myeloid, Acute/genetics , Models, Animal , Myeloproliferative Disorders/genetics
15.
J Hematol Oncol ; 16(1): 19, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36879351

ABSTRACT

BACKGROUND: TP53 mutations, which are present in 5% to 10% of patients with acute myeloid leukemia (AML), are associated with treatment resistance and poor outcomes. First-line therapies for TP53-mutated (TP53m) AML consist of intensive chemotherapy (IC), hypomethylating agents (HMA), or venetoclax combined with HMA (VEN + HMA). METHODS: We conducted a systematic review and meta-analysis to describe and compare treatment outcomes in newly diagnosed treatment-naïve patients with TP53m AML. Randomized controlled trials, single-arm trials, prospective observational studies, and retrospective studies were included that reported on complete remission (CR), CR with incomplete hematologic recovery (CRi), overall survival (OS), event-free survival (EFS), duration of response (DoR), and overall response rate (ORR) among patients with TP53m AML receiving first-line treatment with IC, HMA, or VEN + HMA. RESULTS: Searches of EMBASE and MEDLINE identified 3006 abstracts, and 17 publications describing 12 studies met the inclusion criteria. Random-effects models were used to pool response rates, and time-related outcomes were analyzed with the median of medians method. IC was associated with the greatest CR rate of 43%, and CR rates were 33% for VEN + HMA and 13% for HMA. Rates of CR/CRi were comparable for IC (46%) and VEN + HMA (49%) but were lower for HMA (13%). Median OS was uniformly poor across treatments: IC, 6.5 months; VEN + HMA, 6.2 months; and HMA, 6.1 months. For IC, the EFS estimate was 3.7 months; EFS was not reported for VEN + HMA or HMA. The ORR was 41% for IC, 65% for VEN + HMA, and 47% for HMA. DoR was 3.5 months for IC, 5.0 months for VEN + HMA, and was not reported for HMA. CONCLUSIONS: Despite improved responses seen with IC and VEN + HMA compared to HMA, survival was uniformly poor, and clinical benefits were limited across all treatments for patients with newly diagnosed, treatment-naïve TP53m AML, demonstrating a significant need for improved treatment for this difficult-to-treat population.


Subject(s)
Leukemia, Myeloid, Acute , Humans , Retrospective Studies , Treatment Outcome , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Progression-Free Survival , Mutation , Tumor Suppressor Protein p53/genetics , Observational Studies as Topic
16.
Clin Lymphoma Myeloma Leuk ; 23(2): 127-137, 2023 02.
Article in English | MEDLINE | ID: mdl-36428152

ABSTRACT

BACKGROUND: The global incidence of myelodysplastic syndromes (MDS) has been estimated as 0.06 to 0.26/100,000. Since their introduction, hypomethylating agents have played a central role in the treatment of MDS, with heterogeneous real-world outcomes. MATERIALS AND METHODS: We assessed and synthesized clinical outcomes of azacitidine (AZA) monotherapy in treatment-naïve patients with higher-risk MDS. A systematic literature review was conducted by searching MEDLINE, Embase, and CENTRAL to identify randomized clinical trials (RCTs) and observational studies, both prospective and retrospective, reporting complete remission (CR), partial remission (PR), overall survival (OS), duration of response (DOR), time-to-response (TTR), and myelosuppressive adverse events (AEs) for patients treated with AZA monotherapy. Noncomparative meta-analyses were used to summarize effects. RESULTS: The search identified 3250 abstracts, of which 34 publications describing 16 studies (5 RCTs, 3 prospective, and 8 retrospective observational) were included. Across all studies, pooled CR was 16%; PR was 6%; Median OS was 16.4 months; median DOR was 10.1 months; median TTR was 4.6 months. Proportions of grade 3/4 anemia and thrombocytopenia AEs were 10% and 30%. CONCLUSIONS: The effectiveness and efficacy of AZA monotherapy-as measured by CR and median OS-was limited. These findings highlight a significant unmet medical need for effective treatments for patients with higher-risk MDS.


Subject(s)
Azacitidine , Myelodysplastic Syndromes , Humans , Azacitidine/adverse effects , Antimetabolites, Antineoplastic/adverse effects , Myelodysplastic Syndromes/drug therapy , Treatment Outcome , Remission Induction
17.
Haematologica ; 97(7): 1042-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22315502

ABSTRACT

Juvenile myelomonocytic leukemia is a lethal disease of children characterized by hypersensitivity of hematopoietic progenitors to granulocyte macrophage-colony stimulating factor. Mutations in PTPN11, the gene encoding the protein tyrosine phosphatase Shp2, are common in juvenile myelomonocytic leukemia and induce hyperactivation of the phosphoinositide-3-kinase pathway. We found that genetic disruption of Pik3r1, the gene encoding the Class IA phosphoinositide-3-kinase regulatory subunits p85α, p55α and p50α, significantly reduced hyperproliferation and hyperphosphorylation of Akt in gain-of-function Shp2 E76K-expressing cells. Elevated protein levels of the phosphoinositide-3-kinase catalytic subunit, p110δ, in the Shp2 E76K-expressing Pik3r1-/- cells suggest that p110δ may be a crucial mediator of mutant Shp2-induced phosphoinositide-3-kinase hyperactivation. Consistently, treatment with the p110δ-specific inhibitor, IC87114, or the clinical grade pan-phosphoinositide-3-kinase inhibitor, GDC-0941, reduced granulocyte macrophage-colony stimulating factor hypersensitivity. Treatment with the farnesyltransferase inhibitor, tipifarnib, showed that Shp2 E76K induces hyperactivation of phosphoinositide-3-kinase by both Ras-dependent and Ras-independent mechanisms. Collectively, these findings implicate Class IA phosphoinositide-3-kinase as a relevant molecular target in juvenile myelomonocytic leukemia.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Gene Expression/drug effects , Humans , Leukemia, Myelomonocytic, Juvenile/drug therapy , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/metabolism , Mice , Mutation , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects
18.
Stem Cells ; 29(5): 836-46, 2011 May.
Article in English | MEDLINE | ID: mdl-21374761

ABSTRACT

Wnt/ß-catenin signaling promotes neural differentiation by activation of the neuron-specific transcription factors, Neurogenin1 (Ngn1), NeuroD, and Brn3a, in the nervous system. As neurons in cranial sensory ganglia and dorsal root ganglia transiently express Ngn1, NeuroD, and Brn3a during embryonic development, we hypothesized that Wnt proteins could instructively promote a sensory neuronal fate from mesenchymal stem cells (MSCs) directed to differentiate into neurons. Consistent with our hypothesis, Wnt1 induced expression of sensory neuron markers including Ngn1, NeuroD, and Brn3a, as well as glutamatergic markers in neurally induced MSCs in vitro and promoted engraftment of transplanted MSCs in the inner ear bearing selective loss of sensory neurons in vivo. Given the consensus function of T-cell leukemia 3 (Tlx3), as a glutamatergic selector gene, we postulated that the effects of canonical Wnt signaling on sensory neuron and glutamatergic marker gene expression in MSCs may be mediated by Tlx3. We first confirmed that Wnt1 indeed upregulates Tlx3 expression, which can be suppressed by canonical Wnt inhibitors. Next, our chromatin immunoprecipitation assays revealed that T-cell factor 3/4, Wnt-activated DNA binding proteins, interact with a regulatory region of Tlx3 in MSCs after neural induction. Furthermore, we demonstrated that forced expression of Tlx3 in MSCs induced sensory and glutamatergic neuron markers after neural induction. Together, these results identify Tlx3 as a novel target for canonical Wnt signaling that confers somatic stem cells with a sensory neuron phenotype upon neural induction.


Subject(s)
Cell Differentiation/physiology , Homeodomain Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Wnt1 Protein/metabolism , Cell Differentiation/genetics , Cell Line , Chromatin Immunoprecipitation , DNA Damage/genetics , DNA Damage/physiology , Homeodomain Proteins/genetics , Humans , Immunoblotting , Phagocytosis/genetics , Phagocytosis/physiology , Polymerase Chain Reaction , Telomere/genetics , Wnt1 Protein/genetics
20.
Proc Natl Acad Sci U S A ; 105(15): 5780-5, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18391221

ABSTRACT

The T cell leukemia 3 (Tlx3) gene has been implicated in specification of glutamatergic sensory neurons in the spinal cord. In cranial sensory ganglia, Tlx3 is highly expressed in differentiating neurons during early embryogenesis. To study a role of Tlx3 during neural differentiation, mouse embryonic stem (ES) cells were transfected with a Tlx3 expression vector. ES cells stably expressing Tlx3 were grown in the presence or absence of a neural induction medium. In undifferentiated ES cells, there was no significant difference in gene expression in the presence or absence of Tlx3, even after ES cells were cultured for an extensive time period. In contrast, expression levels of Mash1, Ngn1, and NeuroD were significantly higher in Tlx3-expressing cells after neural induction for 4 days compared with those in cells expressing the control vector. At 7 days after neural induction, whereas expression of the proneural genes was down-regulated, VGLUT2, GluR2, and GluR4 were significantly increased in ES cell-derived neurons expressing Tlx3. The sequential and coordinated expression of the proneural and neuronal subtype-specific genes identifies Tlx3 as a selector gene in ES cells undergoing neural differentiation. In addition, the differential effects of Tlx3 overexpression in undifferentiated ES cells compared with ES cell-derived neurons suggest that Tlx3 exerts context-dependent transcriptional signals on its downstream target genes. The context-dependent function of Tlx3 as a selector gene may be used to establish a novel strategy to conditionally generate excitatory glutamatergic neurons from ES cells to cure various types of neurodegenerative disorders.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Neurons/cytology , Transcription, Genetic , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Glutamic Acid , Homeodomain Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Receptors, AMPA/genetics , Vesicular Glutamate Transport Protein 2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL