Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cytometry A ; 103(10): 763-776, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37421296

ABSTRACT

The bone marrow hematopoietic microenvironment (HME) plays a pivotal role in regulating normal and diseased hematopoiesis. However, the spatial organization of the human HME has not been thoroughly investigated yet. Therefore, we developed a three-dimensional (3D) immunofluorescence model to analyze changes in the cellular architecture in control and diseased bone marrows (BMs). BM biopsies from patients with myeloproliferative neoplasms (MPNs) were stained sequentially for CD31, CD34, CD45, and CD271 with repetitive bleaching steps to realize five color images with DAPI as a nuclear stain. Hematopoietically normal age-matched BM biopsies served as controls. Twelve subsequent slides per sample were stacked to create three-dimensional bone marrow reconstructions with the imaging program Arivis Visions 4D. Iso-surfaces for niche cells and structures were created and exported as mesh objects for spatial distribution analysis in the 3D creation suite Blender. We recapitulated the bone marrow architecture using this approach and produced comprehensive 3D models of endosteal and perivascular BM niches. MPN bone marrows displayed apparent differences compared to the controls, especially concerning CD271 staining density, megakaryocyte (MK) morphology, and distribution. Furthermore, measurements of the spatial relationships of MKs and hematopoietic stem and progenitor cells with vessels and bone structures in their corresponding niche environments revealed the most pronounced differences in the vascular nice in polycythemia vera. Taken together, using a repetitive staining and bleaching approach allowed us to establish a 5-color analysis of human BM biopsies, which is difficult to achieve with conventional staining approaches. Based on this, we generated 3D BM models which recapitulated key pathological features and, importantly, allowed us to define the spatial relationships between different bone marrow cell types. We, therefore, believe that our method can provide new and valuable insights into bone marrow cellular interaction research.

3.
Blood Adv ; 2(23): 3526-3539, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30530780

ABSTRACT

The genetic aberrations that drive mixed phenotype acute leukemia (MPAL) remain largely unknown, with the exception of a small subset of MPALs harboring BCR -ABL1 and MLL translocations. We performed clinicopathologic and genetic evaluation of 52 presumptive MPAL cases at Memorial Sloan Kettering Cancer Center. Only 29 out of 52 (56%) cases were confirmed to be bona fide MPAL according to the 2016 World Heath Organization classification. We identified PHF6 and DNMT3A mutations as the most common recurrent mutations in MPAL, each occurring in 6 out of 26 (23%) cases. These mutations are mutually exclusive of each other and BCR-ABL1/MLL translocations. PHF6- and DNMT3A-mutated MPAL showed marked predilection for T-lineage differentiation (5/6 PHF6 mutated, 6/6 DNMT3A mutated). PHF6-mutated MPAL occurred in a younger patient cohort compared with DNMT3A-mutated cases (median age, 27 years vs 61 years, P < .01). All 3 MPAL cases with both T- and B-lineage differentiation harbored PHF6 mutations. MPAL with T-lineage differentiation was associated with nodal or extramedullary involvement (9/15 [60%] vs 0, P = .001) and a higher relapse incidence (78% vs 22%, P = .017) compared with those without T-lineage differentiation. Sequencing studies on flow-cytometry-sorted populations demonstrated that PHF6 mutations are present in all blast compartments regardless of lineage differentiation with high variant allele frequency, implicating PHF6 as an early mutation in MPAL pathogenesis. In conclusion, PHF6 and DNMT3A mutations are the most common somatic alterations identified in MPAL and appear to define 2 distinct subgroups of MPAL with T-lineage differentiation with inferior outcomes.


Subject(s)
Carrier Proteins/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Leukemia, Biphenotypic, Acute/diagnosis , T-Lymphocytes/cytology , Acute Disease , Adolescent , Adult , Aged , Cell Differentiation/genetics , Child , Child, Preschool , DNA Methyltransferase 3A , Disease-Free Survival , Female , Humans , Infant , Leukemia, Biphenotypic, Acute/genetics , Leukemia, Biphenotypic, Acute/mortality , Male , Middle Aged , Mutation , Repressor Proteins , Survival Rate
4.
Stem Cells Int ; 2018: 3286949, 2018.
Article in English | MEDLINE | ID: mdl-30420889

ABSTRACT

Acute myeloid leukemia (AML) has poor prognosis due to various mutations, e.g., in the FLT3 gene. Therefore, it is important to identify pathways regulated by the activated Flt3 receptor for the discovery of new therapeutic targets. The Myc network of oncogenes and tumor suppressor genes is involved in mechanisms regulating proliferation and survival of cells, including that of the hematopoietic system. In this study, we evaluated the expression of the Myc oncogenes and Mxd antagonists in hematopoietic stem cell and myeloid progenitor populations in the Flt3-ITD-knockin myeloproliferative mouse model. Our data shows that the expression of Myc network genes is changed in Flt3-ITD mice compared with the wild type. Mycn is increased in multipotent progenitors and in the pre-GM compartment of myeloid progenitors in the ITD mice while the expression of several genes in the tumor suppressor Mxd family, including Mxd1, Mxd2, and Mxd4, is concomitantly downregulated, as well as the expression of the Mxd-related gene Mnt and the transcriptional activator Miz-1. LSKCD150+CD48- hematopoietic long-term stem cells are decreased in the Flt3-ITD cells while multipotent progenitors are increased. Of note, PKC412-mediated inhibition of Flt3-ITD signaling results in downregulation of cMyc and upregulation of the Myc antagonists Mxd1, Mxd2, and Mxd4. Our data provides new mechanistic insights into downstream alterations upon aberrant Flt3 signaling and rationale for combination therapies for tyrosine kinase inhibitors with Myc antagonists in treating AML.

5.
Nat Commun ; 9(1): 1770, 2018 05 02.
Article in English | MEDLINE | ID: mdl-29720585

ABSTRACT

Activating signaling mutations are common in acute leukemia with KMT2A (previously MLL) rearrangements (KMT2A-R). These mutations are often subclonal and their biological impact remains unclear. Using a retroviral acute myeloid mouse leukemia model, we demonstrate that FLT3 ITD , FLT3 N676K , and NRAS G12D accelerate KMT2A-MLLT3 leukemia onset. Further, also subclonal FLT3 N676K mutations accelerate disease, possibly by providing stimulatory factors. Herein, we show that one such factor, MIF, promotes survival of mouse KMT2A-MLLT3 leukemia initiating cells. We identify acquired de novo mutations in Braf, Cbl, Kras, and Ptpn11 in KMT2A-MLLT3 leukemia cells that favored clonal expansion. During clonal evolution, we observe serial genetic changes at the Kras G12D locus, consistent with a strong selective advantage of additional Kras G12D . KMT2A-MLLT3 leukemias with signaling mutations enforce Myc and Myb transcriptional modules. Our results provide new insight into the biology of KMT2A-R leukemia with subclonal signaling mutations and highlight the importance of activated signaling as a contributing driver.


Subject(s)
Clonal Evolution , Gene Rearrangement , Histone-Lysine N-Methyltransferase/genetics , Leukemia, Myeloid/genetics , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Acute Disease , Animals , Cell Line, Tumor , Cells, Cultured , Gene Expression Regulation, Leukemic , Leukemia, Myeloid/pathology , Mice, Inbred C57BL , Mice, Transgenic , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins p21(ras)/genetics
6.
Exp Hematol Oncol ; 4: 4, 2015.
Article in English | MEDLINE | ID: mdl-25973343

ABSTRACT

BACKGROUND: Epigenetic code modifications by histone deacetylase inhibitors (HDACi) have been proposed as potential new therapies for lymphoid malignancies. Diffuse large B-cell lymphoma (DLBCL) is the most common type of aggressive lymphoma for which standard first line treatment is the chemotherapy regimen CHOP (cyclophosphamide, doxorubicin, vincristine and prednisone) combined with the monoclonal anti-CD20 antibody rituximab (R-CHOP). The HDACi valproate, which has for long been utilized in anti-convulsive therapy, has been shown to sensitize to chemotherapy in vitro. Valproate upregulates expression of CD20 in lymphoma cell lines; therefore, 48 hour pre-treatment with valproate before first line R-CHOP in DLBCL stages II-IV is evaluated in the phase I clinical trial VALFRID; Valproate as First line therapy in combination with Rituximab and CHOP in Diffuse large B-cell lymphoma. FINDINGS: Pretreatment with valproate at oral doses comparable to anti-convulsive therapy, resulted in upregulation of CD20 mRNA and CD20 protein on the cell surface as measured by qPCR and FACS analysis in lymphoma biopsies from three evaluated patients from the VALFRID study. Valproate-treatment corresponded to increased acetylation of Histone3Lysine9 (H3K9ac) in peripheral blood mononuclear cells (PBMCs), which were employed as surrogate tissue for valproate-related epigenetic modifications. CONCLUSIONS: Valproate treatment at pharmacologically relevant doses resulted in upregulation of CD20 in vivo, and also in expected epigenetic modifications. This suggests that pre-treatment with valproate or other HDACis before anti-CD20 therapy could be advantageous in CD20-low B-cell lymphomas. Further studies are warranted to evaluate this conclusion.

7.
Cell Stem Cell ; 13(5): 535-48, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24054998

ABSTRACT

In jawed vertebrates, development of an adaptive immune-system is essential for protection of the born organism against otherwise life-threatening pathogens. Myeloid cells of the innate immune system are formed early in development, whereas lymphopoiesis has been suggested to initiate much later, following emergence of definitive hematopoietic stem cells (HSCs). Herein, we demonstrate that the embryonic lymphoid commitment process initiates earlier than previously appreciated, prior to emergence of definitive HSCs, through establishment of a previously unrecognized entirely immune-restricted and lymphoid-primed progenitor. Notably, this immune-restricted progenitor appears to first emerge in the yolk sac and contributes physiologically to the establishment of lymphoid and some myeloid components of the immune-system, establishing the lymphomyeloid lineage restriction process as an early and physiologically important lineage-commitment step in mammalian hematopoiesis.


Subject(s)
Hematopoietic Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cells, Cultured , Female , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Hematopoietic Stem Cells/metabolism , Lymphocytes/cytology , Lymphocytes/metabolism , Male , Mice , Myeloid Cells/cytology , Myeloid Cells/metabolism , Polymerase Chain Reaction
8.
Nat Immunol ; 13(4): 412-9, 2012 Feb 19.
Article in English | MEDLINE | ID: mdl-22344248

ABSTRACT

The stepwise commitment from hematopoietic stem cells in the bone marrow to T lymphocyte-restricted progenitors in the thymus represents a paradigm for understanding the requirement for distinct extrinsic cues during different stages of lineage restriction from multipotent to lineage-restricted progenitors. However, the commitment stage at which progenitors migrate from the bone marrow to the thymus remains unclear. Here we provide functional and molecular evidence at the single-cell level that the earliest progenitors in the neonatal thymus had combined granulocyte-monocyte, T lymphocyte and B lymphocyte lineage potential but not megakaryocyte-erythroid lineage potential. These potentials were identical to those of candidate thymus-seeding progenitors in the bone marrow, which were closely related at the molecular level. Our findings establish the distinct lineage-restriction stage at which the T cell lineage-commitment process transits from the bone marrow to the remote thymus.


Subject(s)
B-Lymphocytes/cytology , Cell Lineage/immunology , Lymphoid Progenitor Cells/cytology , Myeloid Cells/cytology , Precursor Cells, B-Lymphoid/cytology , T-Lymphocytes/cytology , Animals , Cell Separation , Flow Cytometry , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Lymphoid Progenitor Cells/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Thymus Gland/cytology
9.
Blood ; 118(13): 3613-21, 2011 Sep 29.
Article in English | MEDLINE | ID: mdl-21813452

ABSTRACT

Acquisition of homozygous activating growth factor receptor mutations might accelerate cancer progression through a simple gene-dosage effect. Internal tandem duplications (ITDs) of FLT3 occur in approximately 25% cases of acute myeloid leukemia and induce ligand-independent constitutive signaling. Homozygous FLT3-ITDs confer an adverse prognosis and are frequently detected at relapse. Using a mouse knockin model of Flt3-internal tandem duplication (Flt3-ITD)-induced myeloproliferation, we herein demonstrate that the enhanced myeloid phenotype and expansion of granulocyte-monocyte and primitive Lin(-)Sca1(+)c-Kit(+) progenitors in Flt3-ITD homozygous mice can in part be mediated through the loss of the second wild-type allele. Further, whereas autocrine FLT3 ligand production has been implicated in FLT3-ITD myeloid malignancies and resistance to FLT3 inhibitors, we demonstrate here that the mouse Flt3(ITD/ITD) myeloid phenotype is FLT3 ligand-independent.


Subject(s)
Gene Dosage/physiology , Gene Duplication/physiology , Loss of Heterozygosity/physiology , Membrane Proteins/genetics , Myeloproliferative Disorders/genetics , fms-Like Tyrosine Kinase 3/physiology , Alleles , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Cells/physiology , Cell Proliferation , Cells, Cultured , Gene Knock-In Techniques , Loss of Heterozygosity/genetics , Male , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Tandem Repeat Sequences/genetics , Tandem Repeat Sequences/physiology , fms-Like Tyrosine Kinase 3/metabolism
10.
Blood ; 118(6): 1544-8, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21628405

ABSTRACT

Lymphoid-primed multipotent progenitors with down-regulated megakaryocyte-erythroid (MkE) potential are restricted to cells with high levels of cell-surface FLT3 expression, whereas HSCs and MkE progenitors lack detectable cell-surface FLT3. These findings are compatible with FLT3 cell-surface expression not being detectable in the fully multipotent stem/progenitor cell compartment in mice. If so, this process could be distinct from human hematopoiesis, in which FLT3 already is expressed in multipotent stem/progenitor cells. The expression pattern of Flt3 (mRNA) and FLT3 (protein) in multipotent progenitors is of considerable relevance for mouse models in which prognostically important Flt3 mutations are expressed under control of the endogenous mouse Flt3 promoter. Herein, we demonstrate that mouse Flt3 expression initiates in fully multipotent progenitors because in addition to lymphoid and granulocyte-monocyte progenitors, FLT3(-) Mk- and E-restricted downstream progenitors are also highly labeled when Flt3-Cre fate mapping is applied.


Subject(s)
Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Multipotent Stem Cells/metabolism , fms-Like Tyrosine Kinase 3/genetics , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Lineage/genetics , Cell Membrane/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Flow Cytometry , Granulocyte Precursor Cells/cytology , Granulocyte Precursor Cells/metabolism , Hematopoietic Stem Cells/cytology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Megakaryocyte Progenitor Cells/cytology , Megakaryocyte Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/cytology , Monocytes/metabolism , Multipotent Stem Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction , fms-Like Tyrosine Kinase 3/metabolism
11.
Blood ; 115(24): 5061-8, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20393130

ABSTRACT

Mice deficient in c-fms-like tyrosine kinase 3 (FLT3) signaling have reductions in early multipotent and lymphoid progenitors, whereas no evident myeloid phenotype has been reported. However, activating mutations of Flt3 are among the most common genetic events in acute myeloid leukemia and mice harboring internal tandem duplications within Flt3 (Flt3-ITD) develop myeloproliferative disease, with characteristic expansion of granulocyte-monocyte (GM) progenitors (GMP), possibly compatible with FLT3-ITD promoting a myeloid fate of multipotent progenitors. Alternatively, FLT3 might be expressed at the earliest stages of GM development. Herein, we investigated the expression, function, and role of FLT3 in recently identified early GMPs. Flt3-cre fate-mapping established that most progenitors and mature progeny of the GM lineage are derived from Flt3-expressing progenitors. A higher expression of FLT3 was found in preGMP compared with GMP, and preGMPs were more responsive to stimulation with FLT3 ligand (FL). Whereas preGMPs and GMPs were reduced in Fl(-/-) mice, megakaryocyte-erythroid progenitors were unaffected and lacked FLT3 expression. Notably, mice deficient in both thrombopoietin (THPO) and FL had a more pronounced GMP phenotype than Thpo(-/-) mice, establishing a role of FL in THPO-dependent and -independent regulation of GMPs, of likely significance for myeloid malignancies with Flt3-ITD mutations.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid, Acute/physiopathology , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism , Animals , Cell Lineage/physiology , Gene Expression/physiology , Granulocytes/cytology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Myeloid Cells/cytology , Signal Transduction/physiology , Thrombopoietin/genetics
12.
Exp Cell Res ; 315(18): 3099-111, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19766114

ABSTRACT

Inhibition of tumor growth factor (TGF)-beta-mediated cell cycle exit is considered an important tumorigenic function of Myc oncoproteins. Here we found that TGF-beta1 enforced G(1) cell cycle arrest and cellular senescence in human U-937 myeloid tumor cells ectopically expressing v-Myc, which contains a stabilizing mutation frequently found in lymphomas. This correlated with induced expression of the Myc antagonist Mad1, resulting in replacement of Myc for Mad1 at target promoters, reduced histone acetylation and strong repression of Myc-driven transcription. The latter was partially reversed by histone deacetylase (HDAC) inhibitors, consistent with involvement of Mad1. Importantly, knockdown of MAD1 expression prevented TGF-beta1-induced senescence, underscoring that Mad1 is a crucial component of this process. Enforced Mad1 expression sensitized U-937-myc cells to TGF-beta and restored phorbol ester-induced cell cycle exit, but could not alone induce G(1) arrest, suggesting that Mad1 is required but not sufficient for cellular senescence. Our results thus demonstrate that TGF-beta can override Myc activity despite a stabilizing cancer mutation and induce senescence in myeloid tumor cells, at least in part by induction of Mad1. TGF-beta-induced senescence, or signals mimicking this pathway, could therefore potentially be explored as a therapeutic principle for treating hematopoietic and other tumors with deregulated MYC expression.


Subject(s)
Cell Cycle Proteins/metabolism , Cellular Senescence , Hematologic Neoplasms/pathology , Myeloid Cells/physiology , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transforming Growth Factor beta1/metabolism , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Line, Tumor , Gene Knockdown Techniques , Hematologic Neoplasms/metabolism , Humans , Myeloid Cells/metabolism , Nuclear Proteins/genetics , RNA, Small Interfering/metabolism
13.
Mol Cancer Ther ; 6(10): 2634-41, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17938259

ABSTRACT

The MYCN protooncogene is involved in the control of cell proliferation, differentiation, and survival of neuroblasts. Deregulation of MYCN by gene amplification contributes to neuroblastoma development and is strongly correlated to advanced disease and poor outcome, emphasizing the urge for new therapeutic strategies targeting MYCN function. The transcription factor N-Myc, encoded by MYCN, regulates numerous genes together with its partner Max, which also functions as a cofactor for the Mad/Mnt family of Myc antagonists/transcriptional repressors. We and others have previously reported that IFN-gamma synergistically potentiates retinoic acid (RA)-induced sympathetic differentiation and growth inhibition in neuroblastoma cells. This study shows that combined treatment of MYCN-amplified neuroblastoma cells with RA+IFN-gamma down-regulates N-Myc protein expression through increased protein turnover, up-regulates Mad1 mRNA and protein, and reduces N-Myc/Max heterodimerization. This results in a shift of occupancy at the ornithine decarboxylase N-Myc/Mad1 target promoter in vivo from N-Myc/Max to Mad1/Max predominance, correlating with histone H4 deacetylation, indicative of a chromatin structure typical of a transcriptionally repressed state. This is further supported by data showing that RA+IFN-gamma treatment strongly represses expression of N-Myc/Mad1 target genes ornithine decarboxylase and hTERT. Our results suggest that combined IFN-gamma and RA signaling can form a basis for new therapeutic strategies targeting N-Myc function for patients with high-risk, MYCN-amplified neuroblastoma.


Subject(s)
Antineoplastic Agents/therapeutic use , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Neuroblastoma/drug therapy , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Acetylation , Blotting, Northern , Blotting, Western , Cell Cycle Proteins/genetics , Cell Differentiation/drug effects , Chromatin/metabolism , Chromatin Immunoprecipitation , Dimerization , Drug Therapy, Combination , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Immunoprecipitation , Interferon-gamma/therapeutic use , N-Myc Proto-Oncogene Protein , Neuroblastoma/metabolism , Neuroblastoma/pathology , Nuclear Proteins/genetics , Ornithine Decarboxylase/genetics , Ornithine Decarboxylase/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/therapeutic use , Tumor Cells, Cultured
14.
Immunity ; 26(4): 407-19, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17433729

ABSTRACT

Recent studies implicated the existence of adult lymphoid-primed multipotent progenitors (LMPPs) with little or no megakaryocyte-erythroid potential, questioning common myeloid and lymphoid progenitors as obligate intermediates in hematopoietic stem cell (HSC) lineage commitment. However, the existence of LMPPs remains contentious. Herein, global and single-cell analyses revealed a hierarchical organization of transcriptional lineage programs, with downregulation of megakaryocyte-erythroid genes from HSCs to LMPPs, sustained granulocyte-monocyte priming, and upregulation of common lymphoid (but not B and T cell-specific) genes. These biological and molecular relationships, implicating almost mutual exclusion of megakaryocyte-erythroid and lymphoid pathways, are established already in fetal hematopoiesis, as evidenced by existence of LMPPs in fetal liver. The identification of LMPPs and hierarchically ordered transcriptional activation and downregulation of distinct lineage programs is compatible with a model for HSC lineage commitment in which the probability for undergoing different lineage commitment fates changes gradually when progressing from HSCs to LMPPs.


Subject(s)
Adult Stem Cells/immunology , Cell Lineage/genetics , Fetus/immunology , Gene Expression Regulation, Developmental , Lymphocytes/immunology , Multipotent Stem Cells/immunology , Adult Stem Cells/cytology , Animals , Fetal Development/genetics , Fetus/cytology , Gene Expression Profiling , Lymphocytes/cytology , Mice , Multipotent Stem Cells/cytology , Transcription, Genetic
15.
Cell ; 121(2): 295-306, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15851035

ABSTRACT

All blood cell lineages derive from a common hematopoietic stem cell (HSC). The current model implicates that the first lineage commitment step of adult pluripotent HSCs results in a strict separation into common lymphoid and common myeloid precursors. We present evidence for a population of cells which, although sustaining a high proliferative and combined lympho-myeloid differentiation potential, have lost the ability to adopt erythroid and megakaryocyte lineage fates. Cells in the Lin-Sca-1+c-kit+ HSC compartment coexpressing high levels of the tyrosine kinase receptor Flt3 sustain granulocyte, monocyte, and B and T cell potentials but in contrast to Lin-Sca-1+c-kit+Flt3- HSCs fail to produce significant erythroid and megakaryocytic progeny. This distinct lineage restriction site is accompanied by downregulation of genes for regulators of erythroid and megakaryocyte development. In agreement with representing a lymphoid primed progenitor, Lin-Sca-1+c-kit+CD34+Flt3+ cells display upregulated IL-7 receptor gene expression. Based on these observations, we propose a revised road map for adult blood lineage development.


Subject(s)
Cell Lineage/physiology , Myeloid Progenitor Cells/classification , Myeloid Progenitor Cells/cytology , Age Factors , Animals , Cell Differentiation , Down-Regulation , Erythrocytes/cytology , Gene Expression Regulation , In Vitro Techniques , Megakaryocytes/cytology , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , fms-Like Tyrosine Kinase 3
16.
Mol Cancer Res ; 2(8): 464-76, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15328373

ABSTRACT

The Mad family proteins are transcriptional repressors belonging to the basic region/helix-loop-helix/leucine zipper family. They share a common obligatory dimerization partner, Max, with the oncoprotein c-Myc and antagonize the function of Myc to activate transcription. The Myc/Max/Mad network has therefore been suggested to function as a molecular switch that regulates cell growth and differentiation by controlling a common set of genes. To study the biological consequences of Mad1 expression for hematopoietic cell growth and differentiation, we used the U-937 monocytic differentiation model to generate cells with inducible Mad1 expression using the reversed tetracycline-controlled transactivator system. The elevated expression of Mad1 in these cells resulted in increased Mad1/Max heterodimer formation correlating with reduced expression of the Myc/Mad target gene ODC. Mad1-expressing U-937 cells in suspension culture proliferated slower and exhibited an increased number of cells in the G1 phase of the cell cycle. Further, growth in semisolid medium was almost completely inhibited. Mad1-expression, however, neither enforced spontaneous differentiation nor enhanced differentiation induced by the phorbol ester 12-O-tetradecanoylphorbol-13-acetate, retinoic acid (RA), or vitamin D3 but rather led to delayed RA-stimulated differentiation. Mad1-expressing cells were further found to be reduced in cell size in all phases of the cells cycle and particularly in response to RA-induced differentiation. Unexpectedly, whereas Fas-induced apoptosis was slightly attenuated in Mad1-expressing U-937 cells, Mad1 sensitized the cells to tumor necrosis factor-alpha-induced apoptosis. These results suggest that Mad1 primarily regulates cell growth and proliferation in these cells, whereas its role in cellular differentiation and survival seems to be more complex.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Differentiation , Monocytes/metabolism , Monocytes/pathology , Nuclear Proteins/metabolism , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Differentiation/drug effects , Cell Proliferation , Cell Survival , Humans , Nuclear Proteins/genetics , Tretinoin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells , fas Receptor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...