Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
J Immunol ; 209(1): 77-92, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35705252

ABSTRACT

The zinc-finger transcription factor GATA-3 plays a crucial role during early T cell development and also dictates later T cell differentiation outcomes. However, its role and collaboration with the Notch signaling pathway in the induction of T lineage specification and commitment have not been fully elucidated. We show that GATA-3 deficiency in mouse hematopoietic progenitors results in an early block in T cell development despite the presence of Notch signals, with a failure to upregulate Bcl11b expression, leading to a diversion along a myeloid, but not a B cell, lineage fate. GATA-3 deficiency in the presence of Notch signaling results in the apoptosis of early T lineage cells, as seen with inhibition of CDK4/6 (cyclin-dependent kinases 4 and 6) function, and dysregulated cyclin-dependent kinase inhibitor 2b (Cdkn2b) expression. We also show that GATA-3 induces Bcl11b, and together with Bcl11b represses Cdkn2b expression; however, loss of Cdkn2b failed to rescue the developmental block of GATA-3-deficient T cell progenitor. Our findings provide a signaling and transcriptional network by which the T lineage program in response to Notch signals is realized.


Subject(s)
GATA3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes , Animals , Cell Differentiation , Cell Lineage , Cyclin-Dependent Kinase Inhibitor Proteins , Gene Regulatory Networks , Mice , Repressor Proteins/genetics , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Tumor Suppressor Proteins/metabolism
2.
Sci Rep ; 11(1): 6777, 2021 03 24.
Article in English | MEDLINE | ID: mdl-33762663

ABSTRACT

Gene expression analysis of individual cells enables characterization of heterogeneous and rare cell populations, yet widespread implementation of existing single-cell gene analysis techniques has been hindered due to limitations in scale, ease, and cost. Here, we present a novel microdroplet-based, one-step reverse-transcriptase polymerase chain reaction (RT-PCR) platform and demonstrate the detection of three targets simultaneously in over 100,000 single cells in a single experiment with a rapid read-out. Our customized reagent cocktail incorporates the bacteriophage T7 gene 2.5 protein to overcome cell lysate-mediated inhibition and allows for one-step RT-PCR of single cells encapsulated in nanoliter droplets. Fluorescent signals indicative of gene expressions are analyzed using a probabilistic deconvolution method to account for ambient RNA and cell doublets and produce single-cell gene signature profiles, as well as predict cell frequencies within heterogeneous samples. We also developed a simulation model to guide experimental design and optimize the accuracy and precision of the assay. Using mixtures of in vitro transcripts and murine cell lines, we demonstrated the detection of single RNA molecules and rare cell populations at a frequency of 0.1%. This low cost, sensitive, and adaptable technique will provide an accessible platform for high throughput single-cell analysis and enable a wide range of research and clinical applications.


Subject(s)
Gene Expression Profiling/methods , Multiplex Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Single-Cell Analysis/methods , Computational Biology/methods , High-Throughput Screening Assays/methods , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/methods , Transcriptome , Workflow
3.
Science ; 345(6203): 1509-12, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25237102

ABSTRACT

The small number of hematopoietic stem and progenitor cells in cord blood units limits their widespread use in human transplant protocols. We identified a family of chemically related small molecules that stimulates the expansion ex vivo of human cord blood cells capable of reconstituting human hematopoiesis for at least 6 months in immunocompromised mice. The potent activity of these newly identified compounds, UM171 being the prototype, is independent of suppression of the aryl hydrocarbon receptor, which targets cells with more-limited regenerative potential. The properties of UM171 make it a potential candidate for hematopoietic stem cell transplantation and gene therapy.


Subject(s)
Fetal Blood/drug effects , Hematopoiesis/drug effects , Hematopoietic Stem Cells/drug effects , Indoles/pharmacology , Pyrimidines/pharmacology , Receptors, Aryl Hydrocarbon/antagonists & inhibitors , Regeneration/drug effects , Animals , Cell Culture Techniques , Fetal Blood/cytology , Fetal Blood/physiology , Genetic Therapy/methods , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/physiology , Humans , Immunocompromised Host , Indoles/chemistry , Mice , Pyrimidines/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
4.
Nat Med ; 20(1): 29-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24292392

ABSTRACT

Tumor recurrence following treatment remains a major clinical challenge. Evidence from xenograft models and human trials indicates selective enrichment of cancer-initiating cells (CICs) in tumors that survive therapy. Together with recent reports showing that CIC gene signatures influence patient survival, these studies predict that targeting self-renewal, the key 'stemness' property unique to CICs, may represent a new paradigm in cancer therapy. Here we demonstrate that tumor formation and, more specifically, human colorectal CIC function are dependent on the canonical self-renewal regulator BMI-1. Downregulation of BMI-1 inhibits the ability of colorectal CICs to self-renew, resulting in the abrogation of their tumorigenic potential. Treatment of primary colorectal cancer xenografts with a small-molecule BMI-1 inhibitor resulted in colorectal CIC loss with long-term and irreversible impairment of tumor growth. Targeting the BMI-1-related self-renewal machinery provides the basis for a new therapeutic approach in the treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms/drug therapy , Heterocyclic Compounds, 2-Ring/pharmacology , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Thiazoles/pharmacology , Animals , Blotting, Western , Bromodeoxyuridine , Cell Line, Tumor , Flow Cytometry , Genetic Vectors/genetics , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Luciferases , Mice, Inbred NOD , Mice, SCID , Polycomb Repressive Complex 1/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Thiazoles/therapeutic use
5.
Proc Natl Acad Sci U S A ; 110(51): 20599-604, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24297922

ABSTRACT

The Lnk (Sh2b3) adaptor protein dampens the response of hematopoietic stem cells and progenitors (HSPCs) to a variety of cytokines by inhibiting JAK2 signaling. As a consequence, Lnk(-/-) mice develop hematopoietic hyperplasia, which progresses to a phenotype resembling the nonacute phase of myeloproliferative neoplasm. In addition, Lnk mutations have been identified in human myeloproliferative neoplasms and acute leukemia. We find that Lnk suppresses the development of radiation-induced acute B-cell malignancies in mice. Lnk-deficient HSPCs recover more effectively from irradiation than their wild-type counterparts, and this resistance of Lnk(-/-) HSPCs to radiation underlies the subsequent emergence of leukemia. A search for the mechanism responsible for radiation resistance identified the cytokine IL-11 as being critical for the ability of Lnk(-/-) HSPCs to recover from irradiation and subsequently become leukemic. In IL-11 signaling, wild-type Lnk suppresses tyrosine phosphorylation of the Src homology region 2 domain-containing phosphatase-2/protein tyrosine phosphatase nonreceptor type 11 and its association with the growth factor receptor-bound protein 2, as well as activation of the Erk MAP kinase pathway. Indeed, Src homology region 2 domain-containing phosphatase-2 has a binding motif for the Lnk Src Homology 2 domain that is phosphorylated in response to IL-11 stimulation. IL-11 therefore drives a pathway that enhances HSPC radioresistance and radiation-induced B-cell malignancies, but is normally attenuated by the inhibitory adaptor Lnk.


Subject(s)
Gamma Rays/adverse effects , Interleukin-11/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leukemia, B-Cell/metabolism , MAP Kinase Signaling System/radiation effects , Neoplasm Proteins/metabolism , Neoplasms, Radiation-Induced/metabolism , Proteins/metabolism , Radiation Tolerance/radiation effects , Adaptor Proteins, Signal Transducing , Amino Acid Motifs , Animals , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Humans , Interleukin-11/genetics , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, B-Cell/genetics , Leukemia, B-Cell/pathology , MAP Kinase Signaling System/genetics , Membrane Proteins , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms, Radiation-Induced/genetics , Neoplasms, Radiation-Induced/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proteins/genetics , Radiation Tolerance/genetics
6.
Cell ; 155(1): 215-27, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24074870

ABSTRACT

Hematopoietic stem cells (HSCs) develop from a specialized subpopulation of endothelial cells known as hemogenic endothelium (HE). Although the HE origin of HSCs is now well established in different species, the signaling pathways that control this transition remain poorly understood. Here, we show that activation of retinoic acid (RA) signaling in aorta-gonad-mesonephros-derived HE ex vivo dramatically enhanced its HSC potential, whereas conditional inactivation of the RA metabolizing enzyme retinal dehydrogenase 2 in VE-cadherin expressing endothelial cells in vivo abrogated HSC development. Wnt signaling completely blocked the HSC inductive effects of RA modulators, whereas inhibition of the pathway promoted the development of HSCs in the absence of RA signaling. Collectively, these findings position RA and Wnt signaling as key regulators of HSC development and in doing so provide molecular insights that will aid in developing strategies for their generation from pluripotent stem cells.


Subject(s)
Hematopoietic Stem Cells/cytology , Tretinoin/metabolism , Aldehyde Oxidoreductases/metabolism , Animals , Aorta/cytology , Aorta/embryology , Down-Regulation , Embryo, Mammalian , Gonads/cytology , Gonads/embryology , Hematopoietic Stem Cells/metabolism , Mesonephros/cytology , Mice , Receptors, Retinoic Acid/metabolism , Wnt Signaling Pathway
7.
Nat Immunol ; 14(10): 1037-44, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23974957

ABSTRACT

The transcription factor GATA-3 is expressed and required for differentiation and function throughout the T lymphocyte lineage. Despite evidence it may also be expressed in multipotent hematopoietic stem cells (HSCs), any role for GATA-3 in these cells has remained unclear. Here we found GATA-3 was in the cytoplasm in quiescent long-term stem cells from steady-state bone marrow but relocated to the nucleus when HSCs cycled. Relocation depended on signaling via the mitogen-activated protein kinase p38 and was associated with a diminished capacity for long-term reconstitution after transfer into irradiated mice. Deletion of Gata3 enhanced the repopulating capacity and augmented the self-renewal of long-term HSCs in cell-autonomous fashion without affecting the cell cycle. Our observations position GATA-3 as a regulator of the balance between self-renewal and differentiation in HSCs that acts downstream of the p38 signaling pathway.


Subject(s)
GATA3 Transcription Factor/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , GATA3 Transcription Factor/genetics , Gene Deletion , Gene Expression , Hematopoiesis/genetics , Hematopoietic Stem Cells/drug effects , Ligands , Mice , Mice, Knockout , Poly I-C/pharmacology , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
PLoS One ; 8(3): e58787, 2013.
Article in English | MEDLINE | ID: mdl-23516556

ABSTRACT

Tachykinins are a large group of neuropeptides with both central and peripheral activity. Despite the increasing number of studies reporting a growth supportive effect of tachykinin peptides in various in vitro stem cell systems, it remains unclear whether these findings are applicable in vivo. To determine how neurokinin-1 receptor (NK-1R) deficient hematopoietic stem cells would behave in a normal in vivo environment, we tested their reconstitution efficiency using competitive bone marrow repopulation assays. We show here that bone marrow taken from NK-1R deficient mice (Tacr1(-/-)) showed lineage specific B and T cell engraftment deficits compared to wild-type competitor bone marrow cells, providing evidence for an involvement of NK-1R signalling in adult hematopoiesis. Tachykinin knockout mice lacking the peptides SP and/or HK-1 (Tac1 (-/-), Tac4 (-/-) and Tac1 (-/-)/Tac4 (-/-) mice) repopulated a lethally irradiated wild-type host with similar efficiency as competing wild-type bone marrow. The difference between peptide and receptor deficient mice indicates a paracrine and/or endocrine mechanism of action rather than autocrine signalling, as tachykinin peptides are supplied by the host environment.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Receptors, Neurokinin-1/metabolism , Signal Transduction , Animals , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Lineage , Female , Gene Knockout Techniques , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Protein Precursors/deficiency , Receptors, Neurokinin-1/deficiency , Receptors, Neurokinin-1/genetics , Substance P/deficiency , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Tachykinins/deficiency
9.
PLoS One ; 8(1): e53161, 2013.
Article in English | MEDLINE | ID: mdl-23326393

ABSTRACT

Hoxb4, a 3'-located Hox gene, enhances hematopoietic stem cell (HSC) activity, while a subset of 5'-located Hox genes is involved in hematopoiesis and leukemogenesis, and some of them are common translocation partners for Nucleoporin 98 (Nup98) in patients with leukemia. Although these Hox gene derivatives are believed to act as transcription regulators, the molecular involvement of the Hox gene derivatives in hematopoiesis and leukemogenesis remains largely elusive. Since we previously showed that Hoxb4 forms a complex with a Roc1-Ddb1-Cul4a ubiquitin ligase core component and functions as an E3 ubiquitin ligase activator for Geminin, we here examined the E3 ubiquitin ligase activities of the 5'-located Hox genes, Hoxa9 and Hoxc13, and Nup98-Hoxa9. Hoxa9 formed a similar complex with the Roc1-Ddb1-Cul4a component to induce ubiquitination of Geminin, but the others did not. Retroviral transduction-mediated overexpression or siRNA-mediated knock-down of Hoxa9 respectively down-regulated or up-regulated Geminin in hematopoietic cells. And Hoxa9 transduction-induced repopulating and clonogenic activities were suppressed by Geminin supertransduction. These findings suggest that Hoxa9 and Hoxb4 differ from Hoxc13 and Nup98-Hoxa9 in their molecular role in hematopoiesis, and that Hoxa9 induces the activity of HSCs and hematopoietic progenitors at least in part through direct down-regulation of Geminin.


Subject(s)
Cell Cycle Proteins/metabolism , Down-Regulation , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Colony-Forming Units Assay/methods , Cullin Proteins/genetics , Cullin Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Geminin , HEK293 Cells , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/genetics , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Protein Binding , RNA Interference , Retroviridae/genetics , Sf9 Cells , Transduction, Genetic , Ubiquitination
10.
Blood ; 119(11): 2510-22, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22174158

ABSTRACT

The stem cell-intrinsic model of self-renewal via asymmetric cell division (ACD) posits that fate determinants be partitioned unequally between daughter cells to either activate or suppress the stemness state. ACD is a purported mechanism by which hematopoietic stem cells (HSCs) self-renew, but definitive evidence for this cellular process remains open to conjecture. To address this issue, we chose 73 candidate genes that function within the cell polarity network to identify potential determinants that may concomitantly alter HSC fate while also exhibiting asymmetric segregation at cell division. Initial gene-expression profiles of polarity candidates showed high and differential expression in both HSCs and leukemia stem cells. Altered HSC fate was assessed by our established in vitro to in vivo screen on a subcohort of candidate polarity genes, which revealed 6 novel positive regulators of HSC function: Ap2a2, Gpsm2, Tmod1, Kif3a, Racgap1, and Ccnb1. Interestingly, live-cell videomicroscopy of the endocytic protein AP2A2 shows instances of asymmetric segregation during HSC/progenitor cell cytokinesis. These results contribute further evidence that ACD is functional in HSC self-renewal, suggest a role for Ap2a2 in HSC activity, and provide a unique opportunity to prospectively analyze progeny from HSC asymmetric divisions.


Subject(s)
Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Asymmetric Cell Division/physiology , Cell Polarity/genetics , Endocytosis/genetics , Hematopoietic Stem Cells/cytology , Neoplastic Stem Cells/pathology , Stem Cells/cytology , Adaptor Protein Complex 2/antagonists & inhibitors , Adaptor Protein Complex 2/genetics , Adaptor Protein Complex alpha Subunits/antagonists & inhibitors , Adaptor Protein Complex alpha Subunits/genetics , Animals , Biomarkers/metabolism , Blotting, Western , Cell Differentiation , Cell Lineage , Cell Proliferation , Flow Cytometry , Gene Expression Profiling , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Leukemia/metabolism , Leukemia/pathology , Mice , Neoplastic Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/physiology
11.
Blood ; 117(16): 4253-61, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21398220

ABSTRACT

Src homology 2 domain-containing phosphatase 2 (Shp2), encoded by Ptpn11, is a member of the nonreceptor protein-tyrosine phosphatase family, and functions in cell survival, proliferation, migration, and differentiation in many tissues. Here we report that loss of Ptpn11 in murine hematopoietic cells leads to bone marrow aplasia and lethality. Mutant mice show rapid loss of hematopoietic stem cells (HSCs) and immature progenitors of all hematopoietic lineages in a gene dosage-dependent and cell-autonomous manner. Ptpn11-deficient HSCs and progenitors undergo apoptosis concomitant with increased Noxa expression. Mutant HSCs/progenitors also show defective Erk and Akt activation in response to stem cell factor and diminished thrombopoietin-evoked Erk activation. Activated Kras alleviates the Ptpn11 requirement for colony formation by progenitors and cytokine/growth factor responsiveness of HSCs, indicating that Ras is functionally downstream of Shp2 in these cells. Thus, Shp2 plays a critical role in controlling the survival and maintenance of HSCs and immature progenitors in vivo.


Subject(s)
Bone Marrow/pathology , Gene Deletion , Hematopoietic Stem Cells/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Animals , Cell Cycle , Cell Death , Epistasis, Genetic , Hematopoietic Stem Cells/cytology , Mice , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Stem Cells/cytology , Stem Cells/metabolism
12.
Blood ; 116(19): 3792-801, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20660792

ABSTRACT

Hemokinin-1 (HK-1), encoded by the TAC4 gene, is a tachykinin peptide that is predominantly expressed in non-neuronal cells, such as immune cells. We have disrupted the mouse TAC4 gene to obtain a better understanding of the actions of HK-1 during hematopoiesis. We demonstrate here that TAC4(-/-) mice exhibit an increase of CD19(+)CD117(+)HSA(+)BP.1(-) "fraction B" pro-B cells in the bone marrow, whereas pre-B, immature, and mature B cells are within the normal range. We show that in vitro cultures derived from TAC4(-/-) bone marrow, sorted "fraction B" pro-B cells or purified long-term reconstituting stem cells, contain significantly higher numbers of pro-B cells compared with controls, suggesting an inhibitory role for HK-1 on developing B cells. Supporting this idea, we show that addition of HK-1 to cultures established from long-term reconstituting stem cells and the newly described intermediate-term reconstituting stem cells leads to a significant decrease of de novo generated pro-B cells. Based on our studies, we postulate that HK-1 plays an inhibitory role in hematopoiesis, and we hypothesize that it may be part of the bone marrow microenvironment that supports and regulates the proliferation and differentiation of hematopoietic cells.


Subject(s)
Lymphopoiesis/genetics , Lymphopoiesis/physiology , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/immunology , Protein Precursors/deficiency , Protein Precursors/genetics , Tachykinins/deficiency , Tachykinins/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA Primers/genetics , Female , Gene Expression , Gene Targeting , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , In Vitro Techniques , Lymphopoiesis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Protein Precursors/immunology , Protein Precursors/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Neurokinin-1/genetics , Tachykinins/immunology , Tachykinins/physiology
13.
Cell Stem Cell ; 7(1): 101-13, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20621054

ABSTRACT

In this study, we describe an in vivo RNA interference functional genetics approach to evaluate the role of 20 different conserved polarity factors and fate determinants in mouse hematopoietic stem cell (HSC) activity. In total, this screen revealed three enhancers and one suppressor of HSC-derived reconstitution. Pard6a, Prkcz, and Msi2 shRNA-mediated depletion significantly impaired HSC repopulation. An in vitro promotion of differentiation was observed after the silencing of these genes, consistent with their function in regulating HSC self-renewal. Conversely, Prox1 knockdown led to in vivo accumulation of primitive and differentiated cells. HSC activity was also enhanced in vitro when Prox1 levels were experimentally reduced, identifying it as a potential antagonist of self-renewal. HSC engineered to overexpress Msi2 or Prox1 showed the reverse phenotype to those transduced with corresponding shRNA vectors. Gene expression profiling studies identified a number of known HSC and cell cycle regulators as potential downstream targets to Msi2 and Prox1.


Subject(s)
Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , RNA Interference/physiology , RNA-Binding Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cells, Cultured , Flow Cytometry , Genetic Vectors/genetics , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Models, Biological , Oligonucleotide Array Sequence Analysis , Protein Kinase C/genetics , Protein Kinase C/metabolism , RNA-Binding Proteins/genetics , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins/genetics
14.
Cell Stem Cell ; 6(1): 48-58, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-20074534

ABSTRACT

Sustained blood cell production depends on divisions by hematopoietic stem cells (HSCs) that yield both differentiating progeny as well as new HSCs via self-renewal. Differentiating progeny remain capable of self-renewal, but only HSCs sustain self-renewal through successive divisions securely enough to maintain clones that persist life-long. Until recently, the first identified next stage consisted of "short-term" reconstituting cells able to sustain clones of differentiating cells for only 4-6 weeks. Here we expand evidence for a numerically dominant "intermediate-term" multipotent HSC stage in mice whose clones persist for 6-8 months before becoming extinct and that are separable from both short-term as well as permanently reconstituting "long-term" HSCs. The findings suggest that the first step in stem cell differentiation consists not in loss of initial capacity for serial self-renewal divisions, but rather in loss of mechanisms that stabilize self-renewing behavior throughout successive future stem cell divisions.


Subject(s)
Cell Differentiation , Cell Division , Hematopoietic Stem Cells/cytology , Animals , Antigens, CD/genetics , Antigens, CD34/genetics , Cell Lineage , Cell Separation , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Integrin alpha2/genetics , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/genetics , Signaling Lymphocytic Activation Molecule Family Member 1 , Time Factors , fms-Like Tyrosine Kinase 3/genetics
15.
Blood ; 115(4): 792-803, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-19850742

ABSTRACT

The majority of long-term reconstituting hematopoietic stem cells (LT-HSCs) in the adult is in G(0), whereas a large proportion of progenitors are more cycling. We show here that the SCL/TAL1 transcription factor is highly expressed in LT-HSCs compared with short-term reconstituting HSCs and progenitors and that SCL negatively regulates the G(0)-G(1) transit of LT-HSCs. Furthermore, when SCL protein levels are decreased by gene targeting or by RNA interference, the reconstitution potential of HSCs is impaired in several transplantation assays. First, the mean stem cell activity of HSCs transplanted at approximately 1 competitive repopulating unit was 2-fold decreased when Scl gene dosage was decreased. Second, Scl(+/-) HSCs were at a marked competitive disadvantage with Scl(+/+) cells when transplanted at 4 competitive repopulating units equivalent. Third, reconstitution of the stem cell pool by adult HSCs expressing Scl-directed shRNAs was decreased compared with controls. At the molecular level, we found that SCL occupies the Cdkn1a and Id1 loci in primary hematopoietic cells and that the expression levels of these 2 regulators of HSC cell cycle and long-term functions are sensitive to Scl gene dosage. Together, our observations suggest that SCL impedes G(0)-G(1) transition in HSCs and regulates their long-term competence.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Animals , Cell Division/drug effects , Cell Division/physiology , Cyclin-Dependent Kinase Inhibitor p21/genetics , G1 Phase/physiology , Gene Expression/physiology , Graft Survival , Hematopoietic Stem Cells/drug effects , Inhibitor of Differentiation Protein 1/genetics , Interleukin-11/pharmacology , Interleukin-6/pharmacology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , RNA Interference , Resting Phase, Cell Cycle/physiology , Stem Cell Factor/pharmacology , T-Cell Acute Lymphocytic Leukemia Protein 1
16.
Proc Natl Acad Sci U S A ; 105(51): 20286-90, 2008 Dec 23.
Article in English | MEDLINE | ID: mdl-19095794

ABSTRACT

The properties and biology of mRNA transcripts can be affected profoundly by the choice of alternative polyadenylation sites, making definition of the 3' ends of transcripts essential for understanding their regulation. Here we show that 22-52% of sequences in commonly used human and murine "full-length" transcript databases may not currently end at bona fide polyadenylation sites. To identify probable transcript termini over the entire murine and human genomes, we analyzed the EST databases for positional clustering of EST ends. The analysis yielded 58,282 murine- and 86,410 human-candidate polyadenylation sites, of which 75% mapped to 23,091 known murine transcripts and 22,891 known human transcripts. The murine dataset correctly predicted 97% of the 3' ends in a manually curated and experimentally supported benchmark transcript set. Of currently known genes, 15% had no associated prediction and 25% had only a single predicted termination site. The remaining genes had an average of 3-4 alternative polyadenylation sites predicted for each murine or human transcript, respectively. The results are made available in the form of tables and an interactive web site that can be mined for rapid assessment of the validity of 3' ends in existing collections, enumeration of potential alternative 3' polyadenylation sites of known transcripts, direct retrieval of terminal sequences for design of probes, and detection of polyadenylation sites not currently mapped to known genes.


Subject(s)
3' Flanking Region , Cluster Analysis , Expressed Sequence Tags , Animals , Humans , Methods , Mice , Poly A , Polyadenylation
17.
Exp Cell Res ; 313(20): 4120-9, 2007 Dec 10.
Article in English | MEDLINE | ID: mdl-17950726

ABSTRACT

Progressive ankylosis (Ank and the human homolog, ANKH) is a transmembrane protein which regulates transport of inorganic pyrophosphate (PPi). ank/ank mice with a mutated ank gene, have calcification and bone ankylosis of the affected joints. In the course of studying these mutant mice, we found that they have microcytosis. These mutant mice have lower mean red blood cell volume (MCV) and lower hemoglobin content in red cells (mean corpuscular hemoglobin, MCH) than normal mice. Using quantitative real-time PCR analysis, we showed that Ank was expressed in the E/Meg bipotent precursor, BFU-E, CFU-E, but there was no Ank expression in the hemoglobinizing erythroblasts. Stable ANKH transfectants in K562 cells highly expressed two immature erythroid cell markers, E-cadherin and endoglin. Enhanced Erythropoietin (Epo) expression and downregulation of SHP-1 were detected in these transfectants. Consequently, the autocrine Epo-EpoR signaling pathway was activated, as evidenced by higher p-Tyr JAK2, p-Tyr EpoR and p-Tyr STAT5B in the ANKH transfectants. Our results revealed a novel function of ANKH in the promotion of early erythroid differentiation in K562 cells. We also showed that ank/ank mice have lower serum levels of Epo than the normal littermates, and this is the likely cause of microcytosis in these mutant mice.


Subject(s)
Cell Differentiation , Endocytosis , Erythroid Cells/cytology , Membrane Proteins/metabolism , Phosphate Transport Proteins/metabolism , Animals , Autocrine Communication , Down-Regulation , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Erythropoietin/blood , Gene Expression Regulation , Humans , Insulin-Like Growth Factor I/metabolism , K562 Cells , Mice , Mice, Mutant Strains , Phosphate Transport Proteins/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Erythropoietin/metabolism , Transfection
18.
Cancer Res ; 65(16): 7151-8, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103065

ABSTRACT

OLIG2 (originally designated BHLHB1) encodes a transcription factor that contains the basic helix-loop-helix motif. Although expression of OLIG2 is normally restricted to neural tissues, overexpression of OLIG2 has been shown in patients with precursor T-cell lymphoblastic lymphoma/leukemia (pre-T LBL). In the current study, we found that overexpression of OLIG2 was not only found in oligodendroglioma samples and normal neural tissue but also in a wide spectrum of malignant cell lines including leukemia, non-small cell lung carcinoma, melanoma, and breast cancer cell lines. To investigate whether enforced expression of OLIG2 is oncogenic, we generated transgenic mice that overexpressed OLIG2 in the thymus. Ectopic OLIG2 expression in the thymus was only weakly oncogenic as only 2 of 85 mice developed pre-T LBL. However, almost 60% of transgenic mice that overexpressed both OLIG2 and LMO1 developed pre-T LBL with large thymic tumor masses. Gene expression profiling of thymic tumors that developed in OLIG2/LMO1 mice revealed up-regulation of Notch1 as well as Deltex1 (Dtx1) and pre T-cell antigen receptor alpha (Ptcra), two genes that are considered to be downstream of Notch1. Of note, we found mutations in the Notch1 heterodimerization or proline-, glutamic acid-, serine-, and threonine-rich domain in three of six primary thymic tumors. In addition, growth of leukemic cell lines established from OLIG2/LMO1 transgenic mice was suppressed by a gamma-secretase inhibitor, suggesting that Notch1 up-regulation is important for the proliferation of OLIG2-LMO1 leukemic cells.


Subject(s)
Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/genetics , Leukemia, T-Cell/genetics , Nerve Tissue Proteins/biosynthesis , Oncogene Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Endopeptidases , Gene Expression Profiling , Humans , LIM Domain Proteins , Leukemia, T-Cell/metabolism , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins , Oligodendrocyte Transcription Factor 2 , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptor, Notch1 , Receptors, Cell Surface/genetics , Thymus Neoplasms/genetics , Thymus Neoplasms/metabolism , Transcription Factors/genetics
19.
Blood ; 105(2): 552-61, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15213094

ABSTRACT

Erythropoietin (EPO) activates many distinct signal transduction cascades on engagement of its receptor. Deletion of the EPO, EPO receptor (EPO-R), or JAK2 genes in mice results in embryonic lethality due to a fatal anemia. EPO activates signal transducer and activator of transcription 1 (STAT1), STAT3, and STAT5a/b transcription factors in erythroid cell lines. Studies have focused on STAT5 as the primary target of EPO-dependent JAK2 activation. However, STAT5a/b(-/-) mice are viable, displaying a nonfatal anemia during embryogenesis, and delayed differentiation in adult erythropoiesis. Importantly, EPO-R cytoplasmic tyrosines are dispensable for viability in vivo. Interestingly, no cytoplasmic tyrosines are required for phosphorylation of STAT1. This led us to examine whether STAT1-deficient mice have altered erythropoiesis. A shift in erythropoiesis was observed in STAT1(-/-) mice, with reduced bone marrow-derived erythroid colony-forming units (CFU-Es) and a compensatory increase in splenic burst-forming units (BFU-Es) and CFU-Es. Both types of splenic-derived cells displayed EPO hyperresponsiveness. A 1.6-fold reduction in total CFU-Es was observed in STAT1-deficient mice, whereas total BFU-Es were comparable. Flow cytometry of STAT1-deficient erythroid cells revealed a less differentiated phenotype, associated with increased apoptosis of early erythroblasts. STAT1-deficient erythroblasts from phenylhydrazine-primed mice displayed enhanced phosphorylation of STAT5a/b, Erk1/2, and protein kinase B (PKB)/Akt. These results illustrate that STAT1 plays an important role in the regulation of erythropoiesis.


Subject(s)
DNA-Binding Proteins/genetics , Erythroid Precursor Cells/physiology , Erythropoiesis/physiology , Trans-Activators/genetics , Anemia/physiopathology , Animals , Apoptosis/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation/physiology , Cell Division/physiology , DNA-Binding Proteins/metabolism , Erythroblasts/enzymology , Erythroid Precursor Cells/cytology , Erythropoietin/pharmacology , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Milk Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , STAT1 Transcription Factor , STAT5 Transcription Factor , Signal Transduction/physiology , Spleen/cytology , Trans-Activators/metabolism
20.
Oncogene ; 23(43): 7223-32, 2004 Sep 20.
Article in English | MEDLINE | ID: mdl-15378082

ABSTRACT

The capacity for sustained self-renewal--the generation of daughter cells having the same regenerative properties as the parent cell--is the defining feature of hematopoietic stem cells (HSCs). Strong evidence exists that self-renewal of HSC is under extrinsic biological control in vivo. A variety of cytokines, morphogenic ligands and associated signaling components influence self-renewal in culture and in vivo. Specific homeobox transcription factors act as powerful intrinsic agonists of HSC self-renewal in vitro and in vivo when supplied either as transduced cDNAs or as externally delivered proteins. These findings provide tools for deepening our knowledge of mechanism and for achievement of clinically useful levels of HSC expansion.


Subject(s)
Hematopoietic Stem Cells/cytology , Animals , Cell Division/drug effects , Cells, Cultured/cytology , Cells, Cultured/drug effects , DNA, Complementary/genetics , Gene Expression Regulation, Developmental , Genes, Homeobox , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Cell Growth Factors/physiology , Hematopoietic Stem Cells/drug effects , Homeodomain Proteins/genetics , Homeodomain Proteins/pharmacology , Homeodomain Proteins/physiology , Humans , Mice , Recombinant Fusion Proteins/physiology , Transcription Factors/genetics , Transcription Factors/pharmacology , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...