Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Nat Rev Cancer ; 23(8): 507, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37353680
2.
Hemasphere ; 6(6): e737, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35647488

ABSTRACT

The hierarchical framework of the adult blood system as we know it from current medical and hematology textbooks, displays a linear branching network of dividing and differentiated cells essential for the growth and maintenance of the healthy organism. This view of the hierarchy has evolved over the last 75 years. An amazing increase in cellular complexity has been realized; however, innovative single-cell technologies continue to uncover essential cell types and functions in animal models and the human blood system. The most potent cell of the hematopoietic hierarchy is the hematopoietic stem cell. Stem cells for adult tissues are the long-lived self-renewing cellular component, which ensure that differentiated tissue-specific cells are maintained and replaced through the entire adult lifespan. Although much blood research is focused on hematopoietic tissue homeostasis, replacement and regeneration during adult life, embryological studies have widened and enriched our understanding of additional developmental hierarchies and interacting cells of this life-sustaining tissue. Here, we review the current state of knowledge of the hierarchical organization and the vast heterogeneity of the hematopoietic system from embryonic to adult stages.

3.
Blood Adv ; 5(3): 829-842, 2021 02 09.
Article in English | MEDLINE | ID: mdl-33560396

ABSTRACT

Integrated molecular signals regulate cell fate decisions in the embryonic aortic endothelium to drive hematopoietic stem cell (HSC) generation during development. The G-protein-coupled receptor 56 (Gpr56, also called Adgrg1) is the most highly upregulated receptor gene in cells that take on hematopoietic fate and is expressed by adult bone marrow HSCs. Despite the requirement for Gpr56 in hematopoietic stem/progenitor cell (HS/PC) generation in zebrafish embryos and the highly upregulated expression of GPR56 in treatment-resistant leukemic patients, its function in normal mammalian hematopoiesis remains unclear. Here, we examine the role of Gpr56 in HS/PC development in Gpr56 conditional knockout (cKO) mouse embryos and Gpr knockout (KO) embryonic stem cell (ESC) hematopoietic differentiation cultures. Our results show a bias toward myeloid differentiation of Gpr56 cKO fetal liver HSCs and an increased definitive myeloid progenitor cell frequency in Gpr56KO ESC differentiation cultures. Surprisingly, we find that mouse Gpr97 can rescue Gpr56 morphant zebrafish hematopoietic generation, and that Gpr97 expression is upregulated in mouse Gpr56 deletion models. When both Gpr56 and Gpr97 are deleted in ESCs, no or few hematopoietic PCs (HPCs) are generated upon ESC differentiation. Together, our results reveal novel and redundant functions for these 2 G-protein coupled receptors in normal mammalian hematopoietic cell development and differentiation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Zebrafish , Animals , Cell Differentiation , Hematopoiesis/genetics , Hematopoietic Stem Cells , Humans , Mice , Receptors, G-Protein-Coupled/genetics , Zebrafish/genetics
4.
Sci Immunol ; 6(56)2021 02 05.
Article in English | MEDLINE | ID: mdl-33547048

ABSTRACT

E-cadherin is a calcium-dependent cell-cell adhesion molecule extensively studied for its involvement in tissue formation, epithelial cell behavior, and suppression of cancer. However, E-cadherin expression in the hematopoietic system has not been fully elucidated. Combining single-cell RNA-sequencing analyses and immunophenotyping, we revealed that progenitors expressing high levels of E-cadherin and contained within the granulocyte-monocyte progenitors (GMPs) fraction have an enriched capacity to differentiate into basophils and mast cells. We detected E-cadherin expression on committed progenitors before the expression of other reported markers of these lineages. We named such progenitors pro-BMPs (pro-basophil and mast cell progenitors). Using RNA sequencing, we observed transcriptional priming of pro-BMPs to the basophil and mast cell lineages. We also showed that GATA-2 directly regulates E-cadherin expression in the basophil and mast cell lineages, thus providing a mechanistic connection between the expression of this cell surface marker and the basophil and mast cell fate specification.


Subject(s)
Cadherins/genetics , GATA2 Transcription Factor/metabolism , Hematopoietic Stem Cells/physiology , Animals , Basophils/physiology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cells, Cultured , Mast Cells/physiology , Mice , Primary Cell Culture , RNA-Seq , Single-Cell Analysis
5.
Development ; 147(23)2020 12 15.
Article in English | MEDLINE | ID: mdl-33323375

ABSTRACT

The central nervous system hosts parenchymal macrophages, known as microglia, and non-parenchymal macrophages, collectively termed border-associated macrophages (BAMs). Microglia, but not BAMs, were reported to be absent in mice lacking a conserved Csf1r enhancer: the fms-intronic regulatory element (FIRE). However, it is unknown whether FIRE deficiency also impacts BAM arrival and/or maintenance. Here, we show that macrophages in the ventricular system of the brain, including Kolmer's epiplexus macrophages, are absent in Csf1rΔFIRE/ΔFIRE mice. Stromal choroid plexus BAMs are also considerably reduced. During normal development, we demonstrate that intracerebroventricular macrophages arrive from embryonic day 10.5, and can traverse ventricular walls in embryonic slice cultures. In Csf1rΔFIRE/ΔFIRE embryos, the arrival of both primitive microglia and intracerebroventricular macrophages was eliminated, whereas the arrival of cephalic mesenchyme and stromal choroid plexus BAMs was only partially restricted. Our results provide new insights into the development and regulation of different CNS macrophage populations.


Subject(s)
Embryonic Development/genetics , Enhancer Elements, Genetic/genetics , Macrophages/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Brain/growth & development , Brain/metabolism , Central Nervous System/growth & development , Embryo, Mammalian , Introns/genetics , Mice , Microglia/metabolism , Parenchymal Tissue/growth & development , Parenchymal Tissue/metabolism , Regulatory Sequences, Nucleic Acid
6.
EMBO J ; 39(8): e104270, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32149421

ABSTRACT

Hematopoietic stem cells (HSCs) develop from the hemogenic endothelium in cluster structures that protrude into the embryonic aortic lumen. Although much is known about the molecular characteristics of the developing hematopoietic cells, we lack a complete understanding of their origin and the three-dimensional organization of the niche. Here, we use advanced live imaging techniques of organotypic slice cultures, clonal analysis, and mathematical modeling to show the two-step process of intra-aortic hematopoietic cluster (IACH) formation. First, a hemogenic progenitor buds up from the endothelium and undergoes division forming the monoclonal core of the IAHC. Next, surrounding hemogenic cells are recruited into the IAHC, increasing their size and heterogeneity. We identified the Notch ligand Dll4 as a negative regulator of the recruitment phase of IAHC. Blocking of Dll4 promotes the entrance of new hemogenic Gfi1+ cells into the IAHC and increases the number of cells that acquire HSC activity. Mathematical modeling based on our data provides estimation of the cluster lifetime and the average recruitment time of hemogenic cells to the cluster under physiologic and Dll4-inhibited conditions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Aorta/embryology , Calcium-Binding Proteins/genetics , Cell Division , Endothelial Progenitor Cells/physiology , Female , Hemangioblasts/physiology , Hematopoietic Stem Cells/physiology , Mice , Mice, Inbred C57BL , Models, Theoretical
7.
Blood ; 134(22): 1929-1940, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31697805

ABSTRACT

Along with the aorta-gonad-mesonephros region, the head is a site of hematopoietic stem and progenitor cell (HS/PC) development in the mouse embryo. Macrophages are present in both these embryonic hemogenic sites, and recent studies indicate a functional interaction of macrophages with hematopoietic cells as they are generated in the aorta. Whereas brain macrophages or "microglia" are known to affect neuronal patterning and vascular circuitry in the embryonic brain, it is unknown whether macrophages play a role in head hematopoiesis. Here, we characterize head macrophages and examine whether they affect the HS/PC output of the hindbrain-branchial arch (HBA) region of the mouse embryo. We show that HBA macrophages are CD45+F4/80+CD11b+Gr1- and express the macrophage-specific Csf1r-GFP reporter. In the HBA of chemokine receptor-deficient (Cx3cr1-/-) embryos, a reduction in erythropoiesis is concomitant with a decrease in HBA macrophage percentages. In cocultures, we show that head macrophages boost hematopoietic progenitor cell numbers from HBA endothelial cells > twofold, and that the proinflammatory factor tumor necrosis factor-α is produced by head macrophages and influences HBA hematopoiesis in vitro. Taken together, head macrophages play a positive role in HBA erythropoiesis and HS/PC expansion and/or maturation, acting as microenvironmental cellular regulators in hematopoietic development.


Subject(s)
Embryo, Mammalian/embryology , Erythropoiesis/physiology , Head/embryology , Hematopoietic Stem Cells/metabolism , Macrophages/metabolism , Animals , Embryo, Mammalian/cytology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Hematopoietic Stem Cells/cytology , Macrophages/cytology , Male , Mice , Mice, Knockout
8.
Cancer Res ; 78(20): 5793-5807, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30154155

ABSTRACT

Combining standard cytotoxic chemotherapy with BCR-ABL1 tyrosine kinase inhibitors (TKI) has greatly improved the upfront treatment of patients with Philadelphia chromosome-positive (Ph+) acute lymphoblastic leukemia (ALL). However, due to the development of drug resistance through both BCR-ABL1-dependent and -independent mechanisms, prognosis remains poor. The STAT5 transcription factor is activated by BCR-ABL1 and by JAK2-dependent cytokine signaling; therefore, inhibiting its activity could address both mechanisms of resistance in Ph+ ALL. We show here that genetic and pharmacologic inhibition of STAT5 activity suppresses cell growth, induces apoptosis, and inhibits leukemogenesis of Ph+ cell lines and patient-derived newly diagnosed and relapsed/TKI-resistant Ph+ ALL cells ex vivo and in mouse models. STAT5 silencing decreased expression of the growth-promoting PIM-1 kinase, the apoptosis inhibitors MCL1 and BCL2, and increased expression of proapoptotic BIM protein. The resulting apoptosis of STAT5-silenced Ph+ BV173 cells was rescued by silencing of BIM or restoration of BCL2 expression. Treatment of Ph+ ALL cells, including samples from relapsed/refractory patients, with the PIM kinase inhibitor AZD1208 and/or the BCL2 family antagonist Sabutoclax markedly suppressed cell growth and leukemogenesis ex vivo and in mice. Together, these studies indicate that targeting STAT5 or STAT5-regulated pathways may provide a new approach for therapy development in Ph+ ALL, especially the relapsed/TKI-resistant disease.Significance: Suppression of STAT5 by BCL2 and PIM kinase inhibitors reduces leukemia burden in mice and constitutes a new potential therapeutic approach against Ph+ ALL, especially in tyrosine kinase inhibitor-resistant disease. Cancer Res; 78(20); 5793-807. ©2018 AACR.


Subject(s)
Gene Expression Regulation, Leukemic , Gene Silencing , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , STAT5 Transcription Factor/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis , Cell Line, Tumor , Cell Survival , Cytokines , Disease Progression , Drug Resistance, Neoplasm , Fusion Proteins, bcr-abl/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mice , Molecular Targeted Therapy , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Recurrence, Local , Neoplasm Transplantation , Philadelphia Chromosome , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/metabolism , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism
9.
J Exp Med ; 215(1): 233-248, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29217535

ABSTRACT

Cell fate is established through coordinated gene expression programs in individual cells. Regulatory networks that include the Gata2 transcription factor play central roles in hematopoietic fate establishment. Although Gata2 is essential to the embryonic development and function of hematopoietic stem cells that form the adult hierarchy, little is known about the in vivo expression dynamics of Gata2 in single cells. Here, we examine Gata2 expression in single aortic cells as they establish hematopoietic fate in Gata2Venus mouse embryos. Time-lapse imaging reveals rapid pulsatile level changes in Gata2 reporter expression in cells undergoing endothelial-to-hematopoietic transition. Moreover, Gata2 reporter pulsatile expression is dramatically altered in Gata2+/- aortic cells, which undergo fewer transitions and are reduced in hematopoietic potential. Our novel finding of dynamic pulsatile expression of Gata2 suggests a highly unstable genetic state in single cells concomitant with their transition to hematopoietic fate. This reinforces the notion that threshold levels of Gata2 influence fate establishment and has implications for transcription factor-related hematologic dysfunctions.


Subject(s)
Cell Differentiation , GATA2 Transcription Factor/genetics , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Single-Cell Analysis , Animals , Female , Fluorescent Antibody Technique , GATA2 Transcription Factor/metabolism , Gene Expression , Genes, Reporter , Male , Mice , Mice, Transgenic , Phenotype , Single-Cell Analysis/methods
10.
Cancer Res ; 78(4): 1097-1109, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29233926

ABSTRACT

Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ ALL) is currently treated with BCR-ABL1 tyrosine kinase inhibitors (TKI) in combination with chemotherapy. However, most patients develop resistance to TKI through BCR-ABL1-dependent and -independent mechanisms. Newly developed TKI can target Ph+ ALL cells with BCR-ABL1-dependent resistance; however, overcoming BCR-ABL1-independent mechanisms of resistance remains challenging because transcription factors, which are difficult to inhibit, are often involved. We show here that (i) the growth of Ph+ ALL cell lines and primary cells is highly dependent on MYB-mediated transcriptional upregulation of CDK6, cyclin D3, and BCL2, and (ii) restoring their expression in MYB-silenced Ph+ ALL cells rescues their impaired proliferation and survival. Levels of MYB and CDK6 were highly correlated in adult Ph+ ALL (P = 0.00008). Moreover, Ph+ ALL cells exhibited a specific requirement for CDK6 but not CDK4 expression, most likely because, in these cells, CDK6 was predominantly localized in the nucleus, whereas CDK4 was almost exclusively cytoplasmic. Consistent with their essential role in Ph+ ALL, pharmacologic inhibition of CDK6 and BCL2 markedly suppressed proliferation, colony formation, and survival of Ph+ ALL cells ex vivo and in mice. In summary, these findings provide a proof-of-principle, rational strategy to target the MYB "addiction" of Ph+ ALL.Significance: MYB blockade can suppress Philadelphia chromosome-positive leukemia in mice, suggesting that this therapeutic strategy may be useful in patients who develop resistance to imatinib and other TKIs used to treat this disease. Cancer Res; 78(4); 1097-109. ©2017 AACR.


Subject(s)
Cyclin-Dependent Kinase 6/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Animals , Cyclin-Dependent Kinase 6/metabolism , Humans , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism
11.
Cell Rep ; 19(2): 295-306, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28402853

ABSTRACT

The role of chromatin structure in lineage commitment of multipotent hematopoietic progenitors (HPCs) is presently unclear. We show here that CD34+ HPCs possess a post-replicative chromatin globally devoid of the repressive histone mark H3K27me3. This H3K27-unmodified chromatin is required for recruitment of lineage-determining transcription factors (TFs) C/EBPα, PU.1, and GATA-1 to DNA just after DNA replication upon cytokine-induced myeloid or erythroid commitment. Blocking DNA replication or increasing H3K27me3 levels prevents recruitment of these TFs to DNA and suppresses cytokine-induced erythroid or myeloid differentiation. However, H3K27me3 is rapidly associated with nascent DNA in more primitive human and murine HPCs. Treatment of these cells with instructive cytokines leads to a significant delay in accumulation of H3K27me3 in nascent chromatin due to activity of the H3K27me3 demethylase UTX. Thus, HPCs utilize special mechanisms of chromatin modification for recruitment of specific TFs to DNA during early stages of lineage specification.


Subject(s)
Cell Differentiation/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/cytology , Jumonji Domain-Containing Histone Demethylases/genetics , Animals , Antigens, CD34/biosynthesis , CCAAT-Enhancer-Binding Protein-alpha/genetics , Cell Lineage/genetics , Chromatin/genetics , DNA Replication/genetics , GATA1 Transcription Factor/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics
12.
Mol Cell ; 66(2): 247-257.e5, 2017 Apr 20.
Article in English | MEDLINE | ID: mdl-28410996

ABSTRACT

Recruitment of transcription factors (TFs) to repressed genes in euchromatin is essential to activate new transcriptional programs during cell differentiation. However, recruitment of all TFs, including pioneer factors, is impeded by condensed H3K27me3-containing chromatin. Single-cell and gene-specific analyses revealed that, during the first hours of induction of differentiation of mammalian embryonic stem cells (ESCs), accumulation of the repressive histone mark H3K27me3 is delayed after DNA replication, indicative of a decondensed chromatin structure in all regions of the replicating genome. This delay provides a critical "window of opportunity" for recruitment of lineage-specific TFs to DNA. Increasing the levels of post-replicative H3K27me3 or preventing S phase entry inhibited recruitment of new TFs to DNA and significantly blocked cell differentiation. These findings suggest that recruitment of lineage-specifying TFs occurs soon after replication and is facilitated by a decondensed chromatin structure. This insight may explain the developmental plasticity of stem cells and facilitate their exploitation for therapeutic purposes.


Subject(s)
Cell Differentiation , Cell Lineage , Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA Replication , DNA/biosynthesis , Embryonic Stem Cells/metabolism , Histones/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , Binding Sites , Cell Plasticity , Chromatin/chemistry , DNA/chemistry , DNA/genetics , DNA Methylation , Gene Expression Regulation, Developmental , Histone Demethylases/metabolism , Histones/chemistry , Humans , Methylation , Mice , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Protein Binding , Structure-Activity Relationship , Time Factors , Transcription Factors/genetics
13.
Oncotarget ; 7(49): 81555-81570, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27835591

ABSTRACT

CML is effectively treated with tyrosine kinase inhibitors (TKIs). However, the efficacy of these drugs is confined to the chronic phase of the disease and development of resistance to TKIs remains a pressing issue. The anti-inflammatory COX2 inhibitor celecoxib has been utilized as anti-tumour drug due to its anti-proliferative activity. However, its effects in hematological malignancies, in particular CML, have not been investigated yet. Thus, we tested biological effects and mechanisms of action of celecoxib in Philadelphia-positive (Ph+) CML and ALL cells.We show here that celecoxib suppresses the growth of Ph+ cell lines by increasing G1-phase and apoptotic cells and reducing S- and G2-phase cells. These effects were independent of COX2 inhibition but required the rapid activation of AMP-activated protein kinase (AMPK) and the consequent inhibition mTORC1 and 2. Treatment with celecoxib also restored GSK3ß function and led to down-regulation of ß-catenin activity through transcriptional and post-translational mechanisms, two effects likely to contribute to Ph+ cell growth suppression by celecoxib.Celecoxib inhibited colony formation of TKI-resistant Ph+ cell lines including those with the T315I BCR-ABL mutation and acted synergistically with imatinib in suppressing colony formation of TKI-sensitive Ph+ cell lines. Finally, it suppressed colony formation of CD34+ cells from CML patients, while sparing most CD34+ progenitors from healthy donors, and induced apoptosis of primary Ph+ ALL cells.Together, these findings indicate that celecoxib may serve as a COX2-independent lead compound to simultaneously target the mTOR and ß-catenin pathways, key players in the resistance of CML stem cells to TKIs.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Celecoxib/pharmacology , Cell Proliferation/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , beta Catenin/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Fusion Proteins, bcr-abl/genetics , Glycogen Synthase Kinase 3 beta/metabolism , HeLa Cells , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction/drug effects , Time Factors , Tumor Cells, Cultured , beta Catenin/genetics
14.
Dev Biol ; 416(1): 34-41, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27235813

ABSTRACT

Hematopoietic cell generation in the midgestation mouse embryo occurs through the natural transdifferentiation of temporally and spatially restricted set of hemogenic endothelial cells. These cells take on hematopoietic fate in the aorta, vitelline and umbilical arteries and appear as hematopoietic cell clusters that emerge from the vascular wall. Genetic and live imaging data have supported this. Recently, the embryonic head has been shown to contain fully functional hematopoietic stem cells (HSC). By lineage tracing, cerebrovascular specific endothelial cells were shown to contribute to the postnatal mouse hematopoietic system. Since Ly6aGFP is a marker of all HSCs, some hematopoietic cluster cells and hemogenic endothelial cells in the midgestation mouse aorta, we examine here whether embryonic head HSCs and vascular endothelial cells are positive for this marker. Whereas some head vasculature, single hematopoietic cells and all HSCs are Ly6aGFP expressing, we do not find clusters of hematopoietic cells emerging from the cerebrovasculature that are characteristic of endothelial-to-hematopoietic transition.


Subject(s)
Antigens, Ly/analysis , Head/embryology , Membrane Proteins/analysis , Animals , Antigens, Differentiation/analysis , Female , Green Fluorescent Proteins , Hematopoietic Stem Cells , Mice , Mice, Inbred C57BL , Mice, Transgenic
15.
Mol Cancer Ther ; 14(8): 1777-93, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26026053

ABSTRACT

Bypassing tyrosine kinases responsible for Stat5a/b phosphorylation would be advantageous for therapy development for Stat5a/b-regulated cancers. Here, we sought to identify small molecule inhibitors of Stat5a/b for lead optimization and therapy development for prostate cancer and Bcr-Abl-driven leukemias. In silico screening of chemical structure databases combined with medicinal chemistry was used for identification of a panel of small molecule inhibitors to block SH2 domain-mediated docking of Stat5a/b to the receptor-kinase complex and subsequent phosphorylation and dimerization. We tested the efficacy of the lead compound IST5-002 in experimental models and patient samples of two known Stat5a/b-driven cancers, prostate cancer and chronic myeloid leukemia (CML). The lead compound inhibitor of Stat5-002 (IST5-002) prevented both Jak2 and Bcr-Abl-mediated phosphorylation and dimerization of Stat5a/b, and selectively inhibited transcriptional activity of Stat5a (IC50 = 1.5µmol/L) and Stat5b (IC50 = 3.5 µmol/L). IST5-002 suppressed nuclear translocation of Stat5a/b, binding to DNA and Stat5a/b target gene expression. IST5-002 induced extensive apoptosis of prostate cancer cells, impaired growth of prostate cancer xenograft tumors, and induced cell death in patient-derived prostate cancers when tested ex vivo in explant organ cultures. Importantly, IST5-002 induced robust apoptotic death not only of imatinib-sensitive but also of imatinib-resistant CML cell lines and primary CML cells from patients. IST5-002 provides a lead structure for further chemical modifications for clinical development for Stat5a/b-driven solid tumors and hematologic malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Prostatic Neoplasms/metabolism , Quantitative Structure-Activity Relationship , STAT5 Transcription Factor/chemistry , Tumor Suppressor Proteins/chemistry , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cluster Analysis , Databases, Factual , Disease Models, Animal , Drug Resistance, Neoplasm , Gene Expression , Gene Expression Profiling , Genes, Reporter , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Male , Mice , Models, Molecular , Molecular Conformation , Phosphorylation , Prostatic Neoplasms/drug therapy , Protein Multimerization , STAT5 Transcription Factor/antagonists & inhibitors , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Small Molecule Libraries , Tissue Culture Techniques , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
16.
Mol Aspects Med ; 39: 102-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23820118

ABSTRACT

Myeloid leukemias are heterogeneous malignancies in morphology, immunophenotype, genetic and epigenetic alterations, and response to therapy. This heterogeneity is thought to depend on the accumulation of secondary mutations enhancing proliferation/survival and/or blocking differentiation in a small subset of leukemia-initiating cells capable of self-renewal. This model of clonal evolution is based on xenotransplantation studies demonstrating that leukemia can be initiated and maintained in immunodeficient mice by a small subset of purified leukemic cells immunophenotypically similar to normal hematopoietic stem cells and is known as the leukemia stem cell model. Since its original formulation, many studies have validated the main conclusion of this model. However, recent data from xenotransplantation studies in more severely immunodeficient mice suggest that imunophenotype and behavior of leukemic stem cells is more heterogeneous and "plastic" than originally thought. We will discuss here the evolution of the leukemia stem cell model and its impact for the therapy of patients with myeloid malignancies.


Subject(s)
Leukemia, Myeloid/pathology , Neoplastic Stem Cells/immunology , Animals , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myeloid/immunology , Mice , Mice, SCID , Neoplasm Transplantation , Neoplastic Stem Cells/pathology
17.
Blood ; 119(9): 2013-23, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22234680

ABSTRACT

Phytohemagglutin-stimulated child and adult leukocytes equally supported CCR5-dependent (R5) and CXCR4-dependent (X4) HIV-1 replication. In contrast, when phytohemagglutin-stimulated leukocytes from either healthy or congenitally immunodeficient children were cultured on feeder cells, they well supported R5, but not X4 HIV-1 replication, whereas both viruses equally spread in adult cells maintained in similar conditions. Both child and adult cells showed similar levels of proliferation and surface expression of CD4, CCR5, CXCR4, CD25, CD69, and HLA-DR. Lack of X4 HIV-1 replication in child versus adult cells was not caused by a differential expression of several known HIV-1 restriction factors. Similar levels of HIV DNA synthesis occurred in child cells infected with R5 and X4 viruses up to 48 hours after infection when R5 HIV-1 showed a significantly superior capacity to spread in culture than X4 virus. Cultured child cells well supported single round vescicular stomatitis virus-G pseudotyped virus replication, whereas superinfection of R5-infected cells with X4 HIV-1 (or vice versa) rescued the replication of this latter virus. Thus, child cells exposed to feeder cell culture represent a novel model system in which the superior capacity of R5 versus X4 viruses to spread can be investigated in primary, untransformed CD4(+) cells.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV-1/immunology , Receptors, CXCR4/metabolism , env Gene Products, Human Immunodeficiency Virus/metabolism , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Line , Cells, Cultured , Child , Child, Preschool , Female , Genetic Therapy , Humans , Infant , Lymphocyte Activation/immunology , Male , Receptors, CCR5/metabolism , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/therapy , Transcription Factors/metabolism , Virus Replication
18.
J Transl Med ; 9 Suppl 1: S8, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21284907

ABSTRACT

Susceptibility to infection by the human immunodeficiency virus type-1 (HIV-1), both in vitro and in vivo, requires the interaction between its envelope (Env) glycoprotein gp120 Env and the primary receptor (R), CD4, and Co-R, either CCR5 or CXCR4, members of the chemokine receptor family. CCR5-dependent (R5) viruses are responsible for both inter-individual transmission and for sustaining the viral pandemics, while CXCR4-using viruses, usually dualtropic R5X4, emerge in ca. 50% of individuals only in the late, immunologically suppressed stage of disease. The hypothesis that such a major biological asymmetry is explained exclusively by the availability of cells expressing CCR5 or CXCR4 is challenged by several evidences. In this regard, binding of the HIV-1 gp120 Env to the entry R complex, i.e. CD4 and a chemokine R, leads to two major events: virion-cell membrane fusion and a cascade of cell signaling. While the fusion/entry process has been well defined, the role of R/Co-R signaling in the HIV-1 life cycle has been less characterized. Indeed, depending on the cellular model studied, the capacity of HIV-1 to trigger a flow of events favoring either its own latency or replication remains a debated issue. In this article, we will review the major findings related to the role of HIV R/Co-R signaling in the steps following viral entry and leading to viral spreading in CD4(+) T lymphocytes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , HIV-1/metabolism , Receptors, Chemokine/metabolism , Animals , Cell Movement , Chemokines/metabolism , Humans , Immune System , Interleukin-4/metabolism , Mutation , Receptors, CCR5/chemistry , Receptors, CXCR4/chemistry , Signal Transduction , Simian Immunodeficiency Virus/metabolism , Th17 Cells/metabolism
19.
Mol Cancer ; 8: 58, 2009 Aug 03.
Article in English | MEDLINE | ID: mdl-19646290

ABSTRACT

BACKGROUND: Akt/PKB is a serine/threonine kinase that has attracted much attention because of its central role in regulating cell proliferation, survival, motility and angiogenesis. Activation of Akt in breast cancer portends aggressive tumour behaviour, resistance to hormone-, chemo-, and radiotherapy-induced apoptosis and it is correlated with decreased overall survival. Recent studies have identified novel tumor-specific substrates of Akt that may provide new diagnostic and prognostic markers and serve as therapeutic targets. This study was undertaken to identify pAkt-interacting proteins and to assess their biological roles in breast cancer cells. RESULTS: We confirmed that one of the pAkt interacting proteins is the Elongation Factor EF1alpha. EF1alpha contains a putative Akt phosphorylation site, but is not phosphorylated by pAkt1 or pAkt2, suggesting that it may function as a modulator of pAkt activity. Indeed, downregulation of EF1alpha expression by siRNAs led to markedly decreased expression of pAkt1 and to less extent of pAkt2 and was associated with reduced proliferation, survival and invasion of HCC1937 cells. Proliferation and survival was further reduced by combining EF1alpha siRNAs with specific pAkt inhibitors whereas EF1alpha downregulation slightly attenuated the decreased invasion induced by Akt inhibitors. CONCLUSION: We show here that EF1alpha is a pAkt-interacting protein which regulates pAkt levels. Since EF1alpha is often overexpressed in breast cancer, the consequences of EF1alpha increased levels for proliferation, survival and invasion will likely depend on the relative concentration of Akt1 and Akt2.


Subject(s)
Breast Neoplasms/metabolism , Peptide Elongation Factor 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Cell Cycle , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoprecipitation , Peptide Elongation Factor 1/antagonists & inhibitors , Peptide Elongation Factor 1/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , RNA Interference
20.
Blood ; 113(8): 1699-709, 2009 Feb 19.
Article in English | MEDLINE | ID: mdl-18941116

ABSTRACT

Urokinase-type plasminogen activator (uPA) signaling via its receptor uPAR inhibits late events in HIV-1 replication in acutely infected primary monocyte-derived macrophages (MDMs) and promonocytic U937 cells. Here we show that U937-derived, chronically infected U1 cells stimulated with phorbol 12-myristate 13-acetate (PMA) express integrins, uPA, and soluble uPAR at levels similar to those of MDMs. uPA inhibited HIV expression in U1 cells incubated with either PMA or tumor necrosis factor-alpha (TNF-alpha), but not with other HIV-inductive cytokines or lipopolysaccharide. Of interest, only PMA and TNF-alpha, but not other HIV-inductive stimuli, induced surface expression of the alpha(M) chain CD11b in U1 cells constitutively expressing CD18, the beta(2) chain of the Mac-1 integrin. Like uPA, fibrinogen, a Mac-1 (CD11b/CD18) ligand, and M25, a peptide homologous to a portion of the beta-propeller region of CD11b preventing its association with uPAR, inhibited HIV virion release in PMA-stimulated U1 cells. Both uPAR small-interference RNA (siRNA) and soluble anti-beta(1)/-beta(2) monoclonal antibodies abolished the anti-HIV effects of uPA, whereas CD11b siRNA reversed the anti-HIV effect of M25, but not that induced by uPA. Thus, either uPA/uPAR interaction, Mac-1 activation, or prevention of its association with uPAR triggers a signaling pathway leading to the inefficient release of HIV from monocytic cells.


Subject(s)
CD11b Antigen/metabolism , CD18 Antigens/metabolism , HIV Infections/metabolism , HIV-1/growth & development , Monocytes/virology , Receptors, Urokinase Plasminogen Activator/metabolism , Activating Transcription Factors/pharmacology , Blood Proteins/pharmacology , Carcinogens/pharmacology , Humans , Integrin beta Chains/metabolism , Ligands , Macrophage-1 Antigen/metabolism , Macrophages/cytology , Macrophages/virology , Monocytes/cytology , Protein Structure, Tertiary , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator/chemistry , Signal Transduction/physiology , Tetradecanoylphorbol Acetate/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells , Up-Regulation/drug effects , Up-Regulation/physiology , Urokinase-Type Plasminogen Activator/metabolism , Virus Replication/drug effects , Virus Replication/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...