Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 76
Filter
1.
Nat Immunol ; 17(8): 956-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27376470

ABSTRACT

During T cell development, multipotent progenitors relinquish competence for other fates and commit to the T cell lineage by turning on Bcl11b, which encodes a transcription factor. To clarify lineage commitment mechanisms, we followed developing T cells at the single-cell level using Bcl11b knock-in fluorescent reporter mice. Notch signaling and Notch-activated transcription factors collaborate to activate Bcl11b expression irrespectively of Notch-dependent proliferation. These inputs work via three distinct, asynchronous mechanisms: an early locus 'poising' function dependent on TCF-1 and GATA-3, a stochastic-permissivity function dependent on Notch signaling, and a separate amplitude-control function dependent on Runx1, a factor already present in multipotent progenitors. Despite their necessity for Bcl11b expression, these inputs act in a stage-specific manner, providing a multitiered mechanism for developmental gene regulation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , GATA3 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 1-alpha/metabolism , Lymphopoiesis/genetics , Receptors, Notch/metabolism , Repressor Proteins/metabolism , T-Lymphocytes/physiology , Tumor Suppressor Proteins/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Tracking , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/genetics , GATA3 Transcription Factor/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Repressor Proteins/genetics , Signal Transduction , Single-Cell Analysis , Tumor Suppressor Proteins/genetics
2.
Blood ; 140(21): 2261-2275, 2022 11 24.
Article in English | MEDLINE | ID: mdl-35605191

ABSTRACT

Adoptive transfer of T cells expressing chimeric antigen receptors (CAR-T) effectively treats refractory hematologic malignancies in a subset of patients but can be limited by poor T-cell expansion and persistence in vivo. Less differentiated T-cell states correlate with the capacity of CAR-T to proliferate and mediate antitumor responses, and interventions that limit tumor-specific T-cell differentiation during ex vivo manufacturing enhance efficacy. NOTCH signaling is involved in fate decisions across diverse cell lineages and in memory CD8+ T cells was reported to upregulate the transcription factor FOXM1, attenuate differentiation, and enhance proliferation and antitumor efficacy in vivo. Here, we used a cell-free culture system to provide an agonistic NOTCH1 signal during naïve CD4+ T-cell activation and CAR-T production and studied the effects on differentiation, transcription factor expression, cytokine production, and responses to tumor. NOTCH1 agonism efficiently induced a stem cell memory phenotype in CAR-T derived from naïve but not memory CD4+ T cells and upregulated expression of AhR and c-MAF, driving heightened production of interleukin-22, interleukin-10, and granzyme B. NOTCH1-agonized CD4+ CAR-T demonstrated enhanced antigen responsiveness and proliferated to strikingly higher frequencies in mice bearing human lymphoma xenografts. NOTCH1-agonized CD4+ CAR-T also provided superior help to cotransferred CD8+ CAR-T, driving improved expansion and curative antitumor responses in vivo at low CAR-T doses. Our data expand the mechanisms by which NOTCH can shape CD4+ T-cell behavior and demonstrate that activating NOTCH1 signaling during genetic modification ex vivo is a potential strategy for enhancing the function of T cells engineered with tumor-targeting receptors.


Subject(s)
Lymphoma , Receptors, Chimeric Antigen , Humans , Mice , Animals , Immunotherapy, Adoptive , CD4-Positive T-Lymphocytes , Transcription Factors , Lymphoma/drug therapy , Receptors, Antigen, T-Cell , Receptor, Notch1/genetics
3.
Blood ; 130(22): 2373-2376, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29021230

ABSTRACT

On 1 September 2017, the US Food and Drug Administration (FDA) approved gemtuzumab ozogamicin (GO) for the treatment of adults with newly diagnosed CD33+ acute myeloid leukemia and for patients aged ≥2 years with CD33+ acute myeloid leukemia who have experienced a relapse or who have not responded to initial treatment. This signals a new chapter in the long and unusual story of GO, which was the first antibody-drug conjugate approved for human use by the FDA.


Subject(s)
Aminoglycosides/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Aminoglycosides/administration & dosage , Aminoglycosides/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Clinical Trials as Topic , Gemtuzumab , Humans , Meta-Analysis as Topic , Sialic Acid Binding Ig-like Lectin 3/analysis
4.
Development ; 140(18): 3799-808, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23924634

ABSTRACT

Genetic regulation of the cell fate transition from lateral plate mesoderm to the specification of cardiomyocytes requires suppression of Wnt/ß-catenin signaling, but the mechanism for this is not well understood. By analyzing gene expression and chromatin dynamics during directed differentiation of human embryonic stem cells (hESCs), we identified a suppressor of Wnt/ß-catenin signaling, transmembrane protein 88 (TMEM88), as a potential regulator of cardiovascular progenitor cell (CVP) specification. During the transition from mesoderm to the CVP, TMEM88 has a chromatin signature of genes that mediate cell fate decisions, and its expression is highly upregulated in advance of key cardiac transcription factors in vitro and in vivo. In early zebrafish embryos, tmem88a is expressed broadly in the lateral plate mesoderm, including the bilateral heart fields. Short hairpin RNA targeting of TMEM88 during hESC cardiac differentiation increases Wnt/ß-catenin signaling, confirming its role as a suppressor of this pathway. TMEM88 knockdown has no effect on NKX2.5 or GATA4 expression, but 80% of genes most highly induced during CVP development have reduced expression, suggesting adoption of a new cell fate. In support of this, analysis of later stage cell differentiation showed that TMEM88 knockdown inhibits cardiomyocyte differentiation and promotes endothelial differentiation. Taken together, TMEM88 is crucial for heart development and acts downstream of GATA factors in the pre-cardiac mesoderm to specify lineage commitment of cardiomyocyte development through inhibition of Wnt/ß-catenin signaling.


Subject(s)
Membrane Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Wnt Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cell Lineage/genetics , Down-Regulation/genetics , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Membrane Proteins/genetics , Mice , Models, Biological , Signal Transduction/genetics , Stem Cells/cytology , Stem Cells/metabolism , Up-Regulation/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics , beta Catenin/metabolism
5.
Blood ; 123(5): 650-8, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24243972

ABSTRACT

Increasing evidence supports the importance of cell extrinsic regulation in stem cell fate control. Hematopoietic stem cells (HSC) are responsive to local signals from their niche and to systemic feedback from progenitors and mature cells. The Notch ligand Delta-1 (DL1), a key component of the stem cell niche, regulates human hematopoietic lineage development in a dose-dependent manner and has been used clinically for primitive progenitor expansion. How DL1 acts to regulate HSC fate and whether these actions are related to its lineage skewing effects are poorly understood. Here we demonstrate that, although DL1 activates signal transducer and activator of transcription 3 signaling similarly to the gp130-activating cytokine interleukin-6 (IL-6), it has opposite effects on myeloid cell production. Mechanistically, these different outcomes are attributable to a DL1-mediated reduction in membrane (m)-bound IL-6 receptor (R) expression, converting progenitor cells from being directly IL-6 responsive to requiring both IL-6 and soluble (s) IL-6R for activation. Concomitant reduction of both mIL-6R (by DL1 supplementation) and sIL-6R (using dynamically fed cultures) reduced myeloid cell production and led to enhanced outputs of human HSCs. This work describes a new mode of cytokine action in which DL1 changes cytokine receptor distributions on hematopoietic cells, altering feedback networks and their impact on stem cell fate.


Subject(s)
Hematopoietic Stem Cells/cytology , Interleukin-6/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Paracrine Communication , Animals , Cells, Cultured , Female , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Janus Kinases/metabolism , Mice , Mice, SCID , STAT3 Transcription Factor/metabolism
6.
Blood ; 121(5): 717-8, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23372149

ABSTRACT

In this issue of Blood, Rafii et al present an elegant study of human embryonic stem cell (ESC)­derived hematopoiesis incorporating live imaging at the single-cell level to track hematopoietic lineage potential during the endothelial to hematopoietic transition.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Transduction, Genetic , Humans
7.
Blood ; 122(7): 1162-73, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23733337

ABSTRACT

Notch signaling regulates several cellular processes including cell fate decisions and proliferation in both invertebrates and mice. However, comparatively less is known about the role of Notch during early human development. Here, we examined the function of Notch signaling during hematopoietic lineage specification from human pluripotent stem cells of both embryonic and adult fibroblast origin. Using immobilized Notch ligands and small interfering RNA to Notch receptors we have demonstrated that Notch1, but not Notch2, activation induced hairy and enhancer of split 1 (HES1) expression and generation of committed hematopoietic progenitors. Using gain- and loss-of-function approaches, this was shown to be attributed to Notch-signaling regulation through HES1, which dictated cell fate decisions from bipotent precursors either to the endothelial or hematopoietic lineages at the clonal level. Our study reveals a previously unappreciated role for the Notch pathway during early human hematopoiesis, whereby Notch signaling via HES1 represents a toggle switch of hematopoietic vs endothelial fate specification.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryonic Stem Cells/cytology , Endothelium, Vascular/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/cytology , Receptor, Notch1/metabolism , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Biomarkers/metabolism , Blotting, Western , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Dermis/cytology , Dermis/metabolism , Embryonic Stem Cells/metabolism , Endothelium, Vascular/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/antagonists & inhibitors , Homeodomain Proteins/genetics , Humans , Immunoenzyme Techniques , Induced Pluripotent Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Small Interfering/genetics , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptors, Notch/metabolism , Signal Transduction , Transcription Factor HES-1
8.
Blood ; 122(8): 1455-63, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23770776

ABSTRACT

Outcomes in acute myeloid leukemia (AML) remain unsatisfactory, and novel treatments are urgently needed. One strategy explores antibodies and their drug conjugates, particularly those targeting CD33. Emerging data with gemtuzumab ozogamicin (GO) demonstrate target validity and activity in some patients with AML, but efficacy is limited by heterogeneous drug conjugation, linker instability, and a high incidence of multidrug resistance. We describe here the development of SGN-CD33A, a humanized anti-CD33 antibody with engineered cysteines conjugated to a highly potent, synthetic DNA cross-linking pyrrolobenzodiazepine dimer via a protease-cleavable linker. The use of engineered cysteine residues at the sites of drug linker attachment results in a drug loading of approximately 2 pyrrolobenzodiazepine dimers per antibody. In preclinical testing, SGN-CD33A is more potent than GO against a panel of AML cell lines and primary AML cells in vitro and in xenotransplantation studies in mice. Unlike GO, antileukemic activity is observed with SGN-CD33A in AML models with the multidrug-resistant phenotype. Mechanistic studies indicate that the cytotoxic effects of SGN-CD33A involve DNA damage with ensuing cell cycle arrest and apoptotic cell death. Together, these data suggest that SGN-CD33A has CD33-directed antitumor activity and support clinical testing of this novel therapeutic in patients with AML.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , Benzodiazepines/chemistry , Drug Resistance, Neoplasm , Immunoconjugates/chemistry , Leukemia, Myeloid, Acute/drug therapy , Sialic Acid Binding Ig-like Lectin 3/chemistry , Animals , Apoptosis , Cell Cycle , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Cysteine/genetics , Dimerization , Drug Design , HEK293 Cells , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/immunology , Mice
9.
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
10.
Blood ; 119(26): 6198-208, 2012 Jun 28.
Article in English | MEDLINE | ID: mdl-22286199

ABSTRACT

Although the identification of cancer stem cells as therapeutic targets is now actively being pursued in many human malignancies, the leukemic stem cells in acute myeloid leukemia (AML) are a paradigm of such a strategy. Heterogeneity of these cells was suggested by clonal analyses indicating the existence of both leukemias resulting from transformed multipotent CD33(-) stem cells as well others arising from, or predominantly involving, committed CD33(+) myeloid precursors. The latter leukemias, which may be associated with an intrinsically better prognosis, offer a particularly attractive target for stem cell-directed therapies. Targeting the CD33 differentiation antigen with gemtuzumab ozogamicin was the first attempt of such an approach. Emerging clinical data indicate that gemtuzumab ozogamicin is efficacious not only for acute promyelocytic leukemia but, in combination with conventional chemotherapy, also for other favorable- and intermediate-risk AMLs, providing the first proof-of-principle evidence for the validity of this strategy. Herein, we review studies on the nature of stem cells in AML, discuss clinical data on the effectiveness of CD33-directed therapy, and consider the mechanistic basis for success and failure in various AML subsets.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , Neoplastic Stem Cells/immunology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Clinical Trials as Topic/statistics & numerical data , Humans , Leukemia, Myeloid, Acute/pathology , Models, Biological , Molecular Targeted Therapy/methods , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Sialic Acid Binding Ig-like Lectin 3 , Treatment Outcome
11.
Blood ; 119(8): 1856-60, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22219225

ABSTRACT

SIRT1 is an NAD(+)-dependent histone deacetylase implicated in the establishment of the primitive hematopoietic system during mouse embryonic development. However, investigation of the role of SIRT1 in adult hematopoiesis has been complicated by the high perinatal mortality of SIRT1-deficient mice (SIRT1(-/-)). We performed a comprehensive in vivo study of the hematopoietic stem cell (HSC) compartment in adult SIRT1(-/-) mice and show that, apart from anemia and leukocytosis in older mice, the production of mature blood cells, lineage distribution within hematopoietic organs, and frequencies of the most primitive HSC populations are comparable to those of wild-type littermate controls. Furthermore, we show that SIRT1-deficient BM cells confer stable long-term reconstitution in competitive repopulation and serial transplantation experiments. The results of the present study rule out an essential physiologic role for cell-autonomous SIRT1 signaling in the maintenance of the adult HSC compartment in mice.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , Sirtuin 1/physiology , Age Factors , Animals , Antigens, CD/metabolism , Antigens, Ly/metabolism , Blood Cell Count , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Female , Flow Cytometry , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Immunophenotyping , Male , Membrane Proteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-kit/metabolism , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Family Member 1 , Sirtuin 1/deficiency , Sirtuin 1/genetics , Time Factors
12.
Blood ; 119(16): 3705-11, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22378848

ABSTRACT

CD33 is expressed on the majority of acute myeloid leukemia (AML) leukemic blasts and is the target for gemtuzumab ozogamicin (GO), a toxin-conjugated anti-CD33 mAb. In the present study, we quantified the CD33 mean fluorescent intensity of leukemic blasts prospectively in 619 de novo pediatric AML patients enrolled in Children's Oncology Group GO-containing clinical trials and determined its correlation with disease characteristics and clinical outcome. CD33 expression varied more than 2-log fold; a median mean fluorescent intensity of 129 (range, 3-1550.07) was observed. Patients were divided into 4 quartiles, quartiles 1-4 (Q1-4) based on expression and disease characteristics and clinical response defined across quartiles. High CD33 expression was associated with high-risk FLT3/ITD mutations (P < .001) and was inversely associated with low-risk disease (P < .001). Complete remission (CR) rates were similar, but patients in Q4 had significantly lower overall survival (57% ± 16% vs 77% ± 7%, P = .002) and disease-free survival from CR (44% ± 16% vs 62% ± 8%, P = .022). In a multivariate model, high CD33 expression remained a significant predictor of overall survival (P = .011) and disease-free survival (P = .038) from CR. Our findings suggest that CD33 expression is heterogeneous within de novo pediatric AML. High expression is associated with adverse disease features and is an independent predictor of inferior outcome. The correlation between CD33 expression and GO response is under investigation. These studies are registered at www.clinicaltrials.gov as NCT00070174 and NCT00372593.


Subject(s)
Aminoglycosides/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antineoplastic Agents/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Child , Child, Preschool , Drug Monitoring/methods , Female , Gemtuzumab , Humans , Infant , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/immunology , Male , Proportional Hazards Models , Prospective Studies , Risk Factors , Sialic Acid Binding Ig-like Lectin 3 , Treatment Outcome , Young Adult
13.
Blood ; 117(23): 6083-90, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21436068

ABSTRACT

Despite progress in our understanding of the growth factors that support the progressive maturation of the various cell lineages of the hematopoietic system, less is known about factors that govern the self-renewal of hematopoietic stem and progenitor cells (HSPCs), and our ability to expand human HSPC numbers ex vivo remains limited. Interest in stem cell expansion has been heightened by the increasing importance of HSCs in the treatment of both malignant and nonmalignant diseases, as well as their use in gene therapy. To date, most attempts to ex vivo expand HSPCs have used hematopoietic growth factors but have not achieved clinically relevant effects. More recent approaches, including our studies in which activation of the Notch signaling pathway has enabled a clinically relevant ex vivo expansion of HSPCs, have led to renewed interest in this arena. Here we briefly review early attempts at ex vivo expansion by cytokine stimulation followed by an examination of our studies investigating the role of Notch signaling in HSPC self-renewal. We will also review other recently developed approaches for ex vivo expansion, primarily focused on the more extensively studied cord blood-derived stem cell. Finally, we discuss some of the challenges still facing this field.


Subject(s)
Cell Culture Techniques/methods , Cytokines/pharmacology , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Fetal Blood/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Receptors, Notch/metabolism , Signal Transduction/drug effects
14.
Blood ; 118(7): 1784-96, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21715312

ABSTRACT

The mechanisms regulating the emergence of BM prothymocytes remain poorly characterized. Genome-wide transcriptome analyses looking for genes expressed in human prothymocytes led to the identification of AF1q/MLLT11 as a candidate gene conceivably involved in this process. Analysis of AF1q protein subcellular localization and intracellular trafficking showed that despite pronounced karyophily, it was subjected to constitutive nuclear export followed by ubiquitin-mediated degradation in the centrosomal area. Using in vitro assays based on either forced expression or shRNA-mediated silencing of AF1q, we provide evidence that the protein promotes T- over B-cell differentiation in multipotent hematopoietic progenitors. At the molecular level, AF1q confers to multipotent progenitors an increased susceptibility to Delta-like/Notch-mediated signaling. Consistent with these findings, enforced AF1q expression in humanized mice fosters the emergence of BM CD34(+)CD7(+) prothymocytes, enhances subsequent thymus colonization, and accelerates intrathymic T-cell development. In contrast, AF1q silencing provokes a global shift of BM lymphopoiesis toward the B-cell lineage, hinders prothymocyte development, inhibits thymus colonization, and leads to intrathymic accumulation of B cells. Our results indicate that AF1q cooperates with the Notch signaling pathway to foster the emergence of BM prothymocytes and drive subsequent intrathymic specification toward the T-cell lineage.


Subject(s)
Hematopoietic Stem Cells/cytology , Lymphopoiesis , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , T-Lymphocytes/cytology , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cells, Cultured , Gene Silencing , HeLa Cells , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, SCID , Molecular Sequence Data , Neoplasm Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins/genetics , Sequence Alignment , Signal Transduction , T-Lymphocytes/metabolism
15.
Blood ; 117(17): 4449-59, 2011 Apr 28.
Article in English | MEDLINE | ID: mdl-21372153

ABSTRACT

Notch signaling critically mediates various hematopoietic lineage decisions and is induced in mammals by Notch ligands that are classified into 2 families, Delta-like (Delta-like-1, -3 and -4) and Jagged (Jagged1 and Jagged2), based on structural homology with both Drosophila ligands Delta and Serrate, respectively. Because the functional differences between mammalian Notch ligands were still unclear, we have investigated their influence on early human hematopoiesis and show that Jagged2 affects hematopoietic lineage decisions very similarly as Delta-like-1 and -4, but very different from Jagged1. OP9 coculture experiments revealed that Jagged2, like Delta-like ligands, induces T-lineage differentiation and inhibits B-cell and myeloid development. However, dose-dependent Notch activation studies, gene expression analysis, and promoter activation assays indicated that Jagged2 is a weaker Notch1-activator compared with the Delta-like ligands, revealing a Notch1 specific signal strength hierarchy for mammalian Notch ligands. Strikingly, Lunatic-Fringe- mediated glycosylation of Notch1 potentiated Notch signaling through Delta-like ligands and also Jagged2, in contrast to Jagged1. Thus, our results reveal a unique role for Jagged1 in preventing the induction of T-lineage differentiation in hematopoietic stem cells and show an unexpected functional similarity between Jagged2 and the Delta-like ligands.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Calcium-Binding Proteins/metabolism , Cell Differentiation/physiology , Cells, Cultured , Drosophila Proteins , Glycosylation , Glycosyltransferases/metabolism , Hematopoietic Stem Cells/metabolism , Humans , Jagged-1 Protein , Jagged-2 Protein , Receptor, Notch1/metabolism , Serrate-Jagged Proteins , Signal Transduction/physiology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
16.
Blood ; 118(17): 4561-6, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21873548

ABSTRACT

IDH1 SNP rs11554137 was recently reported in association with poor prognosis in normal karyotype adult acute myeloid leukemia (AML). We aimed to determine the prevalence, clinical associations, and prognostic significance of SNP rs11554137 in unselected pediatric and adult AML patients. Diagnostic marrow specimens from 527 AML patients treated on the pediatric trial Children's Oncology Group-AAML03P1 (N = 253) or adult SWOG trials (N = 274) were analyzed for the presence of the SNP. SNP rs11554137 was present in 11% of all patients. SNP status had no prognostic impact on survival in pediatric patients. In adult AML, overall survival for SNP-positive patients was 10% versus 18% for SNP-negative patients (P = .44). Among the 142 adults who achieved complete remission, 5-year relapse-free survival was significantly worse for SNP-positive patients (0% vs 25%, P = .0014). However, among adults with normal cytogenetics, FLT3/ITD was present in 90% of SNP-positive patients versus 59% of SNP-negative patients (P = .0053). In multivariate analysis, adjusting for the effects of age, cytogenetics, and FLT3/ITD, the independent prognostic effect of SNP positivity was not statistically significant (hazard ratio = 1.72, P = .18). The clinical profile of SNP-positive patients suggests that SNP rs11554137 may have biologic effects that bear further investigation. The clinical trials in this study are registered at http://www.clinicaltrials.gov as #NCT000707174 and #NCT00899171.


Subject(s)
Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/diagnosis , Polymorphism, Single Nucleotide/physiology , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Clinical Trials as Topic , Female , Humans , Infant , Infant, Newborn , Isocitrate Dehydrogenase/physiology , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/genetics , Male , Medical Oncology/organization & administration , Middle Aged , Mutation, Missense/physiology , Prognosis , Societies, Medical , Young Adult
17.
Stem Cells ; 30(10): 2212-20, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22865615

ABSTRACT

Transplantation of myogenic stem cells possesses great potential for long-term repair of dystrophic muscle. However, a single donor muscle biopsy is unlikely to provide enough cells to effectively transplant the muscle mass of a patient affected by muscular dystrophy. Expansion of cells ex vivo using traditional culture techniques significantly reduces engraftment potential. We hypothesized that activation of Notch signaling during ex vivo expansion would maintain donor cell engraftment potential. In this study, we expanded freshly isolated canine muscle-derived cells on tissue culture plates coated with Delta-1(ext) -IgG to activate Notch signaling or with human IgG as a control. A model of canine-to-murine xenotransplantation was used to quantitatively compare canine muscle cell engraftment and determine whether engrafted donor cells could function as satellite cells in vivo. We show that Delta-1(ext) -IgG inhibited differentiation of canine muscle-derived cells and increased the level of genes normally expressed in myogenic precursors. Moreover, cells expanded on Delta-1(ext) -IgG resulted in a significant increase in the number of donor-derived fibers, as compared to cells expanded on human IgG, reaching engraftment levels similar to freshly isolated cells. Importantly, cells expanded on Delta-1(ext) -IgG engrafted to the recipient satellite cell niche and contributed to further regeneration. A similar strategy of expanding human muscle-derived cells on Notch ligand might facilitate engraftment and muscle regeneration for patients affected with muscular dystrophy.


Subject(s)
Graft Survival , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Receptors, Notch/metabolism , Stem Cells/metabolism , Animals , Cell Communication , Cell Differentiation , Cell Proliferation , Cells, Cultured , Dogs , Humans , Immunoglobulin G/pharmacology , Mice , Mice, SCID , Muscle Cells/cytology , Muscle Cells/transplantation , Muscle, Skeletal/cytology , Regeneration , Signal Transduction , Species Specificity , Stem Cell Transplantation , Stem Cells/cytology , Transplantation, Heterologous
18.
bioRxiv ; 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37333272

ABSTRACT

Decoding the gene regulatory mechanisms mediating self-renewal of hematopoietic stem cells (HSCs) during their amplification in the fetal liver (FL) is relevant for advancing therapeutic applications aiming to expand transplantable HSCs, a long-standing challenge. Here, to explore intrinsic and extrinsic regulation of self-renewal in FL-HSCs at the single cell level, we engineered a culture platform designed to recapitulate the FL endothelial niche, which supports the amplification of serially engraftable HSCs ex vivo. Leveraging this platform in combination with single cell index flow cytometry, serial transplantation assays, and single cell RNA-sequencing, we elucidated previously unrecognized heterogeneity in immunophenotypically defined FL-HSCs and demonstrated that differentiation latency and transcriptional signatures of biosynthetic dormancy are distinguishing properties of self-renewing FL-HSCs with capacity for serial, long-term multilineage hematopoietic reconstitution. Altogether, our findings provide key insights into HSC expansion and generate a novel resource for future exploration of the intrinsic and niche-derived signaling pathways that support FL-HSC self-renewal.

19.
Blood ; 116(26): 5859-66, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-20921340

ABSTRACT

Umbilical cord blood (UCB) is an attractive cell source for hematopoietic cell transplantation (HCT). Here we examine whether the combination of homeobox B4 (HOXB4) and Delta-1 ligand (DL) synergize when used together. Monkey and human UCB CD34(+) cells were transduced with a HOXB4-expressing gammaretroviral vector and cultured with DL. Individual and combined effects of HOXB4 and DL were assessed by colony-forming unit assays, flow cytometry, and nonobese diabetic/severe combined immune deficienct mouse transplantation. The presence of DL yielded higher percentage of CD34(+) and CD7(+) cells and lower percentages of CD14(+) cells than non-DL cultures. Furthermore, HOXB4 yielded higher percentages of CD34(+) and CD14(+) cells than non-HOXB4 cultures. Interestingly, coculture with DL-expressing OP9 cells resulted in better maintenance of HOXB4 than culture in DL-conditioned medium. Culture of HOXB4-transduced human cells in the presence of DL yielded enhanced generation of repopulating cells with higher levels of engraftment of human CD45(+), CD34(+), CD3(+), CD20(+), and CD41(+) cells compared with either factor individually. Our results demonstrate enhanced generation of hematopoietic progenitors by combining HOXB4 and DL; addition of DL further enhances expansion of multipotent cells capable of repopulating lymphoid and megakaryocyte lineages, which is not observed with HOXB4 alone.


Subject(s)
Fetal Blood/cytology , Fetal Blood/transplantation , Homeodomain Proteins/physiology , Membrane Proteins/physiology , Transcription Factors/physiology , Animals , Cells, Cultured , Colony-Forming Units Assay , Culture Media, Conditioned/pharmacology , Fetal Blood/metabolism , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins , Lymphocytes/metabolism , Macaca nemestrina , Mice , Mice, Inbred NOD , Mice, SCID , Myeloid Cells/metabolism , Phenotype
20.
Blood ; 115(4): 757-65, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-19822900

ABSTRACT

We investigated the potential role of an immune reaction in mediating the dominant engraftment of 1 cord blood unit in 14 patients who received a double-unit cord blood transplantation (CBT). In 10 patients, dominant engraftment of a single donor unit emerged by day 28 after CBT. In 9 of these 10 patients, a significant subset of CD8(+) CD45RO(+/-)CCR7(-) T cells, present in peripheral blood mononuclear cells and derived from the engrafting cord blood unit, produced interferon-gamma (IFN-gamma) in response to the nonengrafting unit. No significant population of IFN-gamma-secreting cells was detectable when posttransplantation peripheral blood mononuclear cells were stimulated against cells from the engrafted unit (P < .001) or from a random human leukocyte antigen disparate third party (P = .003). Three patients maintained persistent mixed chimerism after CBT, and no significant IFN-gamma-secreting cells were detected after similar stimulations in these patients (P < .005). Our data provide the first direct evidence in human double-unit CBT recipients that immune rejection mediated by effector CD8(+) T cells developing after CBT from naive precursors is responsible for the failure of 1 unit to engraft. Future investigations based on these findings may result in strategies to predict a dominant unit and enhance graft-versus-leukemia effect.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Cord Blood Stem Cell Transplantation/methods , Graft Rejection/immunology , Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cells/cytology , Leukemia/therapy , Adolescent , Adult , Aged , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation/immunology , Flow Cytometry , Hematopoiesis , Humans , Interferon-gamma/metabolism , Leukemia/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/therapy , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Transplantation Chimera/immunology , Transplantation Conditioning , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL