Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Mar 29.
Article in English | MEDLINE | ID: mdl-37034724

ABSTRACT

Transition between activation and quiescence programs in hematopoietic stem and progenitor cells (HSC/HSPCs) is perceived to be governed intrinsically and by microenvironmental co-adaptation. However, HSC programs dictating both transition and adaptability, remain poorly defined. Single cell multiome analysis divulging differential transcriptional activity between distinct HSPC states, indicated for the exclusive absence of Fli-1 motif from quiescent HSCs. We reveal that Fli-1 activity is essential for HSCs during regenerative hematopoiesis. Fli-1 directs activation programs while manipulating cellular sensory and output machineries, enabling HSPCs co-adoptability with a stimulated vascular niche. During regenerative conditions, Fli-1 presets and enables propagation of niche-derived Notch1 signaling. Constitutively induced Notch1 signaling is sufficient to recuperate functional HSC impairments in the absence of Fli-1. Applying FLI-1 modified-mRNA transduction into lethargic adult human mobilized HSPCs, enables their vigorous niche-mediated expansion along with superior engraftment capacities. Thus, decryption of stem cell activation programs offers valuable insights for immune regenerative medicine.

2.
Sci Rep ; 11(1): 6104, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33731767

ABSTRACT

The human airway epithelium lining the bronchial tree contains basal cells that proliferate, differentiate, and communicate with other components of their microenvironment. One method that cells use for intercellular communication involves the secretion of exosomes and other extracellular vesicles (EVs). We isolated exosome-enriched EVs that were produced from an immortalized human airway basal cell line (BCi-NS1.1) and found that their secretion is increased by exposure to cigarette smoke extract, suggesting that this stress stimulates release of EVs which could affect signaling to other cells. We have previously shown that primary human airway basal cells secrete vascular endothelial growth factor A (VEGFA) which can activate MAPK signaling cascades in endothelial cells via VEGF receptor-2 (VEGFR2). Here, we show that exposure of endothelial cells to exosome-enriched airway basal cell EVs promotes the survival of these cells and that this effect also involves VEGFR2 activation and is, at least in part, mediated by VEGFA present in the EVs. These observations demonstrate that EVs are involved in the intercellular signaling between airway basal cells and the endothelium which we previously reported. The downstream signaling pathways involved may be distinct and specific to the EVs, however, as increased phosphorylation of Akt, STAT3, p44/42 MAPK, and p38 MAPK was not seen following exposure of endothelial cells to airway basal cell EVs.


Subject(s)
Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , MAP Kinase Signaling System , Tobacco Products , Tobacco Smoke Pollution , Cell Line, Transformed , Endothelial Cells/pathology , Extracellular Vesicles/pathology , Humans , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
4.
J Immunol ; 199(8): 2701-2712, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28931604

ABSTRACT

Total body irradiation (TBI) damages hematopoietic cells in the bone marrow and thymus; however, the long-term effects of irradiation with aging remain unclear. In this study, we found that the impact of radiation on thymopoiesis in mice varied by sex and dose but, overall, thymopoiesis remained suppressed for ≥12 mo after a single exposure. Male and female mice showed a long-term dose-dependent reduction in thymic cKit+ lymphoid progenitors that was maintained throughout life. Damage to hematopoietic stem cells (HSCs) in the bone marrow was dose dependent, with as little as 0.5 Gy causing a significant long-term reduction. In addition, the potential for T lineage commitment was radiation sensitive with aging. Overall, the impact of irradiation on the hematopoietic lineage was more severe in females. In contrast, the rate of decline in thymic epithelial cell numbers with age was radiation-sensitive only in males, and other characteristics including Ccl25 transcription were unaffected. Taken together, these data suggest that long-term suppression of thymopoiesis after sublethal irradiation was primarily due to fewer progenitors in the BM combined with reduced potential for T lineage commitment. A single irradiation dose also caused synchronization of thymopoiesis, with a periodic thymocyte differentiation profile persisting for at least 12 mo postirradiation. This study suggests that the number and capability of HSCs for T cell production can be dramatically and permanently damaged after a single relatively low TBI dose, accelerating aging-associated thymic involution. Our findings may impact evaluation and therapeutic intervention of human TBI events.


Subject(s)
Bone Marrow Cells/physiology , Hematopoiesis/radiation effects , Immunologic Deficiency Syndromes/immunology , Lymphoid Progenitor Cells/physiology , T-Lymphocytes/physiology , Thymus Gland/radiation effects , Aging , Animals , Cell Differentiation , Cell Lineage , Cells, Cultured , Female , Immunologic Deficiency Syndromes/etiology , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-kit/metabolism , Thymus Gland/immunology , Whole-Body Irradiation/adverse effects
5.
Oncotarget ; 7(28): 43062-43075, 2016 Jul 12.
Article in English | MEDLINE | ID: mdl-27248664

ABSTRACT

DNA damaging agents cause rapid shrinkage of tumors and form the basis of chemotherapy for sarcomas despite significant toxicities. Drugs having superior efficacy and wider therapeutic windows are needed to improve patient outcomes. We used cell proliferation and apoptosis assays in sarcoma cell lines and benign cells; γ-H2AX expression, comet assay, immunoblot analyses and drug combination studies in vitro and in patient derived xenograft (PDX) models. BO-1055 caused apoptosis and cell death in a concentration and time dependent manner in sarcoma cell lines. BO-1055 had potent activity (submicromolar IC50) against Ewing sarcoma and rhabdomyosarcoma, intermediate activity in DSRCT (IC50 = 2-3µM) and very weak activity in osteosarcoma (IC50 >10µM) cell lines. BO-1055 exhibited a wide therapeutic window compared to other DNA damaging drugs. BO-1055 induced more DNA double strand breaks and γH2AX expression in cancer cells compared to benign cells. BO-1055 showed inhibition of tumor growth in A673 xenografts and caused tumor regression in cyclophosphamide resistant patient-derived Ewing sarcoma xenografts and A204 xenografts. Combination of BO-1055 and irinotecan demonstrated synergism in Ewing sarcoma PDX models. Potent activity on sarcoma cells and its relative lack of toxicity presents a strong rationale for further development of BO-1055 as a therapeutic agent.


Subject(s)
DNA Damage , Nitrogen Mustard Compounds/pharmacology , Phenylurea Compounds/pharmacology , Sarcoma/drug therapy , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Synergism , HCT116 Cells , Humans , Irinotecan , MCF-7 Cells , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nitrogen Mustard Compounds/administration & dosage , Phenylurea Compounds/administration & dosage , Sarcoma/genetics , Sarcoma/pathology
6.
Article in English | MEDLINE | ID: mdl-27169377

ABSTRACT

Accumulated DNA damage in hematopoietic stem cells is a primary mechanism of aging-associated dysfunction in human hematopoiesis. About 70 years ago, atomic-bomb (A-bomb) radiation induced DNA damage and functional decreases in the hematopoietic system of A-bomb survivors in a radiation dose-dependent manner. The peripheral blood cell populations then recovered to a normal range, but accompanying cells derived from hematopoietic stem cells still remain that bear molecular changes possibly caused by past radiation exposure and aging. In the present study, we evaluated radiation-related changes in the frequency of phosphorylated (Ser-139) H2AX (γH2AX) foci formation in circulating CD34-positive/lineage marker-negative (CD34+Lin-) hematopoietic stem and progenitor cells (HSPCs) among 226Hiroshima A-bomb survivors. An association between the frequency of γH2AX foci formation in HSPCs and the radiation dose was observed, but the γH2AX foci frequency was not significantly elevated by past radiation. We found a negative correlation between the frequency of γH2AX foci formation and the length of granulocyte telomeres. A negative interaction effect between the radiation dose and the frequency of γH2AX foci was suggested in a proportion of a subset of HSPCs as assessed by the cobblestone area-forming cell assay (CAFC), indicating that the self-renewability of HSPCs may decrease in survivors who were exposed to a higher radiation dose and who had more DNA damage in their HSPCs. Thus, although many years after radiation exposure and with advancing age, the effect of DNA damage on the self-renewability of HSPCs may be modified by A-bomb radiation exposure.


Subject(s)
Hematopoietic Stem Cells/cytology , Stem Cells/cytology , Age Factors , Aged , Aged, 80 and over , Cell Differentiation/genetics , Cell Differentiation/physiology , DNA Damage/genetics , DNA Damage/physiology , Hematopoietic Stem Cells/metabolism , Histones/genetics , Histones/metabolism , Humans , Middle Aged , Stem Cells/metabolism
7.
Radiat Res ; 185(1): 69-76, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26720799

ABSTRACT

It is not yet known whether hematopoietic stem and progenitor cells (HSPCs) are compromised in the aging population of atomic bomb (A-bomb) survivors after their exposure nearly 70 years ago. To address this, we evaluated age- and radiation-related changes in different subtypes of circulating HSPCs among the CD34-positive/lineage marker-negative (CD34(+)Lin(-)) cell population in 231 Hiroshima A-bomb survivors. We enumerated functional HSPC subtypes, including: cobblestone area-forming cells; long-term culture-initiating cells; erythroid burst-forming units; granulocyte and macrophage colony-forming units; and T-cell and natural killer cell progenitors using cell culture. We obtained the count of each HSPC subtype per unit volume of blood and the proportion of each HSPC subtype in CD34(+)Lin(-) cells to represent the lineage commitment trend. Multivariate analyses, using sex, age and radiation dose as variables, showed significantly decreased counts with age in the total CD34(+)Lin(-) cell population and all HSPC subtypes. As for the proportion, only T-cell progenitors decreased significantly with age, suggesting that the commitment to the T-cell lineage in HSPCs continuously declines with age throughout the lifetime. However, neither the CD34(+)Lin(-) cell population, nor HSPC subtypes showed significant radiation-induced dose-dependent changes in counts or proportions. Moreover, the correlations of the proportions among HSPC subtypes in the survivors properly revealed the hierarchy of lineage commitments. Taken together, our findings suggest that many years after exposure to radiation and with advancing age, the number and function of HSPCs in living survivors as a whole may have recovered to normal levels.


Subject(s)
Blood Cells/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/radiation effects , Nuclear Weapons/statistics & numerical data , Radiation Exposure/statistics & numerical data , Survivors/statistics & numerical data , Age Distribution , Aged , Aged, 80 and over , Blood Cells/radiation effects , Cell Proliferation/radiation effects , Dose-Response Relationship, Radiation , Female , Humans , Japan/epidemiology , Male , Sex Distribution
8.
J Am Chem Soc ; 137(40): 13167-75, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26401918

ABSTRACT

Human granulocyte colony-stimulating factor (G-CSF) is an endogenous glycoprotein involved in hematopoiesis. Natively glycosylated and nonglycosylated recombinant forms, lenograstim and filgrastim, respectively, are used clinically to manage neutropenia in patients undergoing chemotherapeutic treatment. Despite their comparable therapeutic potential, the purpose of O-linked glycosylation at Thr133 remains a subject of controversy. In light of this, we have developed a synthetic platform to prepare G-CSF aglycone with the goal of enabling access to native and designed glycoforms with site-selectivity and glycan homogeneity. To address the synthesis of a relatively large, aggregation-prone sequence, we advanced an isonitrile-mediated ligation method. The chemoselective activation and coupling of C-terminal peptidyl Gly thioacids with the N-terminus of an unprotected peptide provide ligated peptides directly in a manner complementary to that with conventional native chemical ligation-desulfurization strategies. Herein, we describe the details and application of this method as it enabled the convergent total synthesis of G-CSF aglycone.


Subject(s)
Granulocyte Colony-Stimulating Factor/chemistry , Peptides/chemistry , Amino Acid Sequence , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data
9.
Top Curr Chem ; 362: 1-26, 2015.
Article in English | MEDLINE | ID: mdl-25805144

ABSTRACT

Glycoproteins are an important class of naturally occurring biomolecules which play a pivotal role in many biological processes. They are biosynthesized as complex mixtures of glycoforms through post-translational protein glycosylation. This fact, together with the challenges associated with producing them in homogeneous form, has hampered detailed structure-function studies of glycoproteins as well as their full exploitation as potential therapeutic agents. By contrast, chemical synthesis offers the unique opportunity to gain access to homogeneous glycoprotein samples for rigorous biological evaluation. Herein, we review recent methods for the assembly of complex glycopeptides and glycoproteins and present several examples from our laboratory towards the total chemical synthesis of clinically relevant glycosylated proteins that have enabled synthetic access to full-length homogeneous glycoproteins.


Subject(s)
Glycoproteins/chemical synthesis , Glycosylation , Molecular Structure , Protein Engineering
10.
Proc Natl Acad Sci U S A ; 111(8): 2885-90, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24516138

ABSTRACT

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a medicinally important glycoprotein, used as an immunostimulant following bone-marrow transplant. On the basis of reports of its potential utility as an anticancer vaccine adjuvant, we undertook to develop a synthetic route toward single-glycoform GM-CSF. We describe herein a convergent total synthesis of GM-CSF aglycone and two homogeneous glycoforms. Analytical and biological studies confirm the structure and activity of these synthetic congeners.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/chemical synthesis , Models, Molecular , Protein Conformation , Alanine/chemistry , Amino Acid Sequence , Cysteine/chemistry , Escherichia coli , Glycosylation , Molecular Sequence Data , Molecular Structure
11.
Science ; 342(6164): 1357-1360, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24337294

ABSTRACT

Erythropoietin is a signaling glycoprotein that controls the fundamental process of erythropoiesis, orchestrating the production and maintenance of red blood cells. As administrated clinically, erythropoietin has a polypeptide backbone with complex dishomogeneity in its carbohydrate domains. Here we describe the total synthesis of homogeneous erythropoietin with consensus carbohydrate domains incorporated at all of the native glycosylation sites. The oligosaccharide sectors were built by total synthesis and attached stereospecifically to peptidyl fragments of the wild-type primary sequence, themselves obtained by solid-phase peptide synthesis. The glycopeptidyl constructs were joined by chemical ligation, followed by metal-free dethiylation, and subsequently folded. This homogeneous erythropoietin glycosylated at the three wild-type aspartates with N-linked high-mannose sialic acid-containing oligosaccharides and O-linked glycophorin exhibits Procrit-level in vivo activity in mice.


Subject(s)
Erythropoietin/administration & dosage , Erythropoietin/chemical synthesis , Amino Acid Sequence , Animals , Aspartic Acid/chemistry , Cells, Cultured , Consensus Sequence , Dose-Response Relationship, Drug , Erythrocyte Count , Erythropoietin/chemistry , Glycophorins/chemistry , Glycosylation , Injections, Subcutaneous , Mannose/chemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , N-Acetylneuraminic Acid/chemistry , Oligosaccharides/chemistry , Reticulocytes/drug effects
12.
Nat Chem Biol ; 9(12): 840-848, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24161946

ABSTRACT

Efforts to develop more effective therapies for acute leukemia may benefit from high-throughput screening systems that reflect the complex physiology of the disease, including leukemia stem cells (LSCs) and supportive interactions with the bone marrow microenvironment. The therapeutic targeting of LSCs is challenging because LSCs are highly similar to normal hematopoietic stem and progenitor cells (HSPCs) and are protected by stromal cells in vivo. We screened 14,718 compounds in a leukemia-stroma co-culture system for inhibition of cobblestone formation, a cellular behavior associated with stem-cell function. Among those compounds that inhibited malignant cells but spared HSPCs was the cholesterol-lowering drug lovastatin. Lovastatin showed anti-LSC activity in vitro and in an in vivo bone marrow transplantation model. Mechanistic studies demonstrated that the effect was on target, via inhibition of HMG-CoA reductase. These results illustrate the power of merging physiologically relevant models with high-throughput screening.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor/methods , Leukemia , Neoplastic Stem Cells/drug effects , Cell Line, Tumor , Hematopoietic Stem Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lovastatin/pharmacology , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/physiology
13.
Blood ; 121(20): 4082-9, 2013 May 16.
Article in English | MEDLINE | ID: mdl-23520338

ABSTRACT

Direct transduction of the homeobox (HOX) protein HOXB4 promotes the proliferation of hematopoietic stem cells (HSCs) without induction of leukemogenesis, but requires frequent administration to overcome its short protein half-life (∼1 hour). We demonstrate here that HOXB4 protein levels are post-translationally regulated by the CUL4 ubiquitin ligase, and define the degradation signal sequence (degron) of HOXB4 required for CUL4-mediated destruction. Additional HOX paralogs share the conserved degron in the homeodomain and are also subject to CUL4-mediated degradation, indicating that CUL4 likely controls the stability of all HOX proteins. Moreover, we engineered a degradation-resistant HOXB4 that conferred a growth advantage over wild-type HOXB4 in myeloid progenitor cells. Direct transduction of recombinant degradation-resistant HOXB4 protein to human adult HSCs significantly enhanced their maintenance in a more primitive state both in vitro and in transplanted NOD/SCID/IL2R-γ(null) mice compared with transduction with wild-type HOXB4 protein. Our studies demonstrate the feasibility of engineering a stable HOXB4 variant to overcome a major technical hurdle in the ex vivo expansion of adult HSCs and early progenitors for human therapeutic use.


Subject(s)
Adult Stem Cells/physiology , Cell Proliferation , Cullin Proteins/physiology , Hematopoietic Stem Cells/physiology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Adult , Adult Stem Cells/metabolism , Animals , Cell Culture Techniques/methods , Cells, Cultured , Cullin Proteins/genetics , Cullin Proteins/metabolism , Feasibility Studies , HeLa Cells , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Primary Cell Culture/methods , Protein Engineering , Proteolysis , Transcription Factors/genetics , Transcription Factors/physiology
14.
Nat Cell Biol ; 15(3): 309-16, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23434824

ABSTRACT

Coordinating the balance between haematopoietic stem cell (HSC) quiescence and self-renewal is crucial for maintaining haematopoiesis lifelong. Equally important for haematopoietic function is modulating HSC localization within the bone marrow niches, as maintenance of HSC function is tightly controlled by a complex network of intrinsic molecular mechanisms and extrinsic signalling interactions with their surrounding microenvironment. In this study we demonstrate that nuclear factor erythroid 2-related factor 2 (Nfe2l2, or Nrf2), well established as a global regulator of the oxidative stress response, plays a regulatory role in several aspects of HSC homeostasis. Nrf2 deficiency results in an expansion of the haematopoietic stem and progenitor cell compartment due to cell-intrinsic hyperproliferation, which was accomplished at the expense of HSC quiescence and self-renewal. We further show that Nrf2 modulates both migration and retention of HSCs in their niche. Moreover, we identify a previously unrecognized link between Nrf2 and CXCR4, contributing, at least partially, to the maintenance of HSC function.


Subject(s)
Bone Marrow/metabolism , Cell Communication , Cell Proliferation , Hematopoiesis/physiology , Hematopoietic Stem Cells/physiology , NF-E2-Related Factor 2/physiology , Stromal Cells/metabolism , Animals , Blotting, Western , Bone Marrow Transplantation , Chromatin Immunoprecipitation , Female , Flow Cytometry , Hematopoietic Stem Cells/cytology , Luciferases/metabolism , Mice , Mice, Knockout , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stromal Cells/cytology , Transfection
15.
Proc Natl Acad Sci U S A ; 109(34): E2276-83, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22652566

ABSTRACT

Most gastrointestinal stromal tumors (GISTs) harbor a gain-of-function mutation in the Kit receptor. GIST patients treated with the tyrosine kinase inhibitor imatinib frequently develop imatinib resistance as a result of second-site Kit mutations. To investigate the consequences of second-site Kit mutations on GIST development and imatinib sensitivity, we engineered a mouse model carrying in the endogenous Kit locus both the Kit(V558Δ) mutation found in a familial case of GIST and the Kit(T669I) (human KIT(T670I)) "gatekeeper" mutation found in imatinib-resistant GIST patients. Similar to Kit(V558/+) mice, Kit(V558;T669I/+) mice developed gastric and colonic interstitial cell of Cajal hyperplasia as well as cecal GIST. In contrast to the single-mutant Kit(V558/+) control mice, treatment of the Kit(V558;T669I/+) mice with either imatinib or dasatinib failed to inhibit oncogenic Kit signaling and GIST growth. However, this resistance could be overcome by treatment of Kit(V558;T669I/+) mice with sunitinib or sorafenib. Although tumor lesions were smaller in Kit(V558;T669I/+) mice than in single-mutant mice, both interstitial cell of Cajal hyperplasia and mast cell hyperplasia were exacerbated in Kit(V558;T669I/+) mice. Strikingly, the Kit(V558;T669I/+) mice developed a pronounced polycythemia vera-like erythrocytosis in conjunction with microcytosis. This mouse model should be useful for preclinical studies of drug candidates designed to overcome imatinib resistance in GIST and to investigate the consequences of oncogenic KIT signaling in hematopoietic as well as other cell lineages.


Subject(s)
Erythrocytes/cytology , Gastrointestinal Stromal Tumors/genetics , Mutation , Piperazines/pharmacology , Polycythemia/genetics , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Benzamides , Cell Lineage , Dasatinib , Disease Models, Animal , Drug Resistance , Drug Resistance, Neoplasm/genetics , Exons , Gastrointestinal Stromal Tumors/drug therapy , Imatinib Mesylate , Mice , Phenotype , Proto-Oncogene Proteins c-kit/metabolism , Thiazoles/pharmacology
16.
J Exp Med ; 208(9): 1757-65, 2011 Aug 29.
Article in English | MEDLINE | ID: mdl-21844206

ABSTRACT

Hematopoietic stem cells (HSCs) self-renew to maintain the lifelong production of all blood populations. Here, we show that the proliferating cell nuclear antigen-associated factor (Paf) is highly expressed in cycling bone marrow HSCs and plays a critical role in hematopoiesis. Mice lacking Paf exhibited reduced bone marrow cellularity; reduced numbers of HSCs and committed progenitors; and leukopenia. These phenotypes are caused by a cell-intrinsic blockage in the development of long-term (LT)-HSCs into multipotent progenitors and preferential loss of lymphoid progenitors caused by markedly increased p53-mediated apoptosis. In addition, LT-HSCs from Paf(-/-) mice had increased levels of reactive oxygen species (ROS), failed to maintain quiescence, and were unable to support LT hematopoiesis. The loss of lymphoid progenitors was likely due the increased levels of ROS in LT-HSCs caused by treatment of Paf(-/-) mice with the anti-oxidant N-acetylcysteine restored lymphoid progenitor numbers to that of Paf(+/+) mice. Collectively, our studies identify Paf as a novel and essential regulator of early hematopoiesis.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , Hematopoiesis/physiology , Lymphoid Progenitor Cells/metabolism , Oncogene Proteins/metabolism , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Carrier Proteins/genetics , Free Radical Scavengers/pharmacology , Hematopoiesis/drug effects , Leukopenia/genetics , Leukopenia/metabolism , Mice , Mice, Knockout , Oncogene Proteins/genetics , Reactive Oxygen Species/metabolism , Time Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Stem Cells ; 28(2): 308-17, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19967789

ABSTRACT

Hematopoietic stem cells (HSCs) can remain quiescent or they can enter the cell cycle, and either self-renew or differentiate. Although cyclin C and cyclin dependent kinase (cdk3) are essential for the transition from the G(0) to the G(1) phase of the cell cycle in human fibroblasts, the role of cyclin C in hematopoietic stem/progenitor cells (HSPCs) is not clear. We have identified an important role of cyclin C (CCNC) in regulating human HSPC quiescence, as knocking down CCNC expression in human cord blood CD34(+) cells resulted in a significant increase in quiescent cells that maintain CD34 expression. CCNC knockdown also promotes in vitro HSPC expansion and enhances their engraftment potential in sublethally irradiated immunodeficient mice. Our studies establish cyclin C as a critical regulator of the G(0)/G(1) transition of human HSPCs and suggest that modulating cyclin C levels may be useful for HSC expansion and more efficient engraftment.


Subject(s)
Cyclin C/physiology , Hematopoietic Stem Cells/physiology , Animals , Antigens, CD34/metabolism , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Division/genetics , Cell Division/physiology , Cells, Cultured , Cyclin C/genetics , Cyclin C/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Ki-67 Antigen/metabolism , Mice , Mice, Inbred NOD , Polymerase Chain Reaction , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology
18.
Cell Stem Cell ; 1(3): 346-52, 2007 Sep 13.
Article in English | MEDLINE | ID: mdl-18371368

ABSTRACT

Parthenogenesis and somatic cell nuclear transfer (SCNT) are two methods for deriving embryonic stem (ES) cells that are genetically matched to the oocyte donor or somatic cell donor, respectively. Using genome-wide single nucleotide polymorphism (SNP) analysis, we demonstrate distinct signatures of genetic recombination that distinguish parthenogenetic ES cells from those generated by SCNT. We applied SNP analysis to the human ES cell line SCNT-hES-1, previously claimed to have been derived by SCNT, and present evidence that it represents a human parthenogenetic ES cell line. Genome-wide SNP analysis represents a means to validate the genetic provenance of an ES cell line.


Subject(s)
Embryonic Stem Cells/metabolism , Nuclear Transfer Techniques , Parthenogenesis , Recombination, Genetic/genetics , Animals , Cell Line , Cytogenetic Analysis , DNA Methylation , Genome, Human/genetics , Heterozygote , Homozygote , Humans , Mice , Polymorphism, Single Nucleotide/genetics , Sequence Analysis, DNA , Specimen Handling
19.
Cancer Res ; 66(24): 11781-91, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17178874

ABSTRACT

The t(7;11)(p15;p15) translocation, observed in acute myelogenous leukemia and myelodysplastic syndrome, generates a chimeric gene where the 5' portion of the sequence encoding the human nucleoporin NUP98 protein is fused to the 3' region of HOXA9. Here, we show that retroviral-mediated enforced expression of the NUP98-HOXA9 fusion protein in cord blood-derived CD34(+) cells confers a proliferative advantage in both cytokine-stimulated suspension cultures and stromal coculture. This advantage is reflected in the selective expansion of hematopoietic stem cells as measured in vitro by cobblestone area-forming cell assays and in vivo by competitive repopulation of nonobese diabetic/severe combined immunodeficient mice. NUP98-HOXA9 expression inhibited erythroid progenitor differentiation and delayed neutrophil maturation in transduced progenitors but strongly enhanced their serial replating efficiency. Analysis of the transcriptosome of transduced cells revealed up-regulation of several homeobox genes of the A and B cluster as well as of Meis1 and Pim-1 and down-modulation of globin genes and of CAAT/enhancer binding protein alpha. The latter gene, when coexpressed with NUP98-HOXA9, reversed the enhanced proliferation of transduced CD34(+) cells. Unlike HOXA9, the NUP98-HOXA9 fusion was protected from ubiquitination mediated by Cullin-4A and subsequent proteasome-dependent degradation. The resulting protein stabilization may contribute to the leukemogenic activity of the fusion protein.


Subject(s)
Antigens, CD34/physiology , Homeodomain Proteins/genetics , Mutant Chimeric Proteins/genetics , Nuclear Pore Complex Proteins/genetics , Stem Cells/cytology , Antigens, CD/physiology , Cell Division , Cell Line , Chromosome Mapping , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 7 , Cloning, Molecular , Colony-Forming Units Assay , Fetal Blood , Gene Fusion , Genetic Vectors , Humans , Oligonucleotide Array Sequence Analysis , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Translocation, Genetic , Umbilical Cord
20.
Methods Enzymol ; 418: 208-42, 2006.
Article in English | MEDLINE | ID: mdl-17141038

ABSTRACT

Murine embryonic stem cells (mESC) readily form embryoid bodies (EBs) that exhibit hematopoietic differentiation. Methods based on EB formation or ESC coculture with murine bone marrow stromal cell lines have revealed pathways of both primitive and definitive hematopoietic differentiation progressing from primitive mesoderm via hemangioblasts to endothelium and hematopoietic stem and progenitor cells. The addition of specific hematopoietic growth factors and morphogens to these cultures enhances the generation of neutrophils, macrophages, megakaryocyte/platelets, and hemoglobinized mature red cells. In addition, selective culture systems have been developed to support differentiation into mature T lymphocytes, natural killer cells, B cells, and dendritic cells. In most cases, culture systems have been developed that support equivalent differentiation of various human ESC (hESC). The major obstacle to translation of ESC hematopoietic cultures to clinical relevance has been the general inability to produce hematopoietic stem cells (HSC) that can engraft adult, irradiated recipients. In this context, the pattern of ES hematopoietic development mirrors the yolk sac phase of hematopoiesis that precedes the appearance of engraftable HSC in the aorta-gonad-mesonephros region. Genetic manipulation of mESC hematopoietic progeny by upregulation of HOXB4 or STAT5 has led to greatly enhanced long- or short-term multilineage hematopoietic engraftment, suggesting that genetic or epigenetic manipulation of these pathways may lead to functional HSC generation from hESC.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Animals , Cell Culture Techniques/methods , Cell Differentiation/physiology , Chorion/cytology , Chorion/physiology , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/physiology , Gene Expression , Hematopoiesis/genetics , Humans , Mice , Primates , Stromal Cells/cytology , Stromal Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...