Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Obes Sci Pract ; 5(3): 203-219, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31275594

ABSTRACT

INTRODUCTION: This report presents a synopsis of a three-part, cross-sector, seminar series held at the George Washington University (GWU) in Washington, DC from February-April, 2018. The overarching goal of the seminar series was to provide a neutral forum for diverse stakeholders to discuss and critically evaluate approaches to address added sugar intake, with a key focus on the role of low-calorie sweeteners (LCS). METHODS: During three seminars, twelve speakers from academic institutions, federal agencies, non-profit organizations, and the food and beverage industries participated in six interactive panel discussions to address: 1) Do Farm Bill Policies Impact Population Sugar Intake? 2) What is the Impact of Sugar-sweetened Beverage (SSB) Taxes on Health and Business? 3) Is Sugar Addictive? 4) Product Reformulation Efforts: Progress, Challenges, and Concerns? 5) Low-calorie Sweeteners: Helpful or Harmful, and 6) Are Novel Sweeteners a Plausible Solution? Discussion of each topic involved brief 15-minute presentations from the speakers, which were followed by a 25-minute panel discussion moderated by GWU faculty members and addressed questions generated by the audience. Sessions were designed to represent opposing views and stimulate meaningful debate. Given the provocative nature of the seminar series, attendee questions were gathered anonymously using Pigeonhole™, an interactive, online, question and answer platform. RESULTS: This report summarizes each presentation and recapitulates key perspectives offered by the speakers and moderators. CONCLUSIONS: The seminar series set the foundation for robust cross-sector dialogue necessary to inform meaningful future research, and ultimately, effective policies for lowering added sugar intakes.

2.
Int J Obes (Lond) ; 40(10): 1523-1528, 2016 10.
Article in English | MEDLINE | ID: mdl-27460603

ABSTRACT

BACKGROUND/OBJECTIVES: State-specific obesity prevalence data are critical to public health efforts to address the childhood obesity epidemic. However, few states administer objectively measured body mass index (BMI) surveillance programs. This study reports state-specific childhood obesity prevalence by age and sex correcting for parent-reported child height and weight bias. SUBJECTS/METHODS: As part of the Childhood Obesity Intervention Cost Effectiveness Study (CHOICES), we developed childhood obesity prevalence estimates for states for the period 2005-2010 using data from the 2010 US Census and American Community Survey (ACS), 2003-2004 and 2007-2008 National Survey of Children's Health (NSCH) (n=133 213), and 2005-2010 National Health and Nutrition Examination Surveys (NHANES) (n=9377; ages 2-17). Measured height and weight data from NHANES were used to correct parent-report bias in NSCH using a non-parametric statistical matching algorithm. Model estimates were validated against surveillance data from five states (AR, FL, MA, PA and TN) that conduct censuses of children across a range of grades. RESULTS: Parent-reported height and weight resulted in the largest overestimation of childhood obesity in males ages 2-5 years (NSCH: 42.36% vs NHANES: 11.44%). The CHOICES model estimates for this group (12.81%) and for all age and sex categories were not statistically different from NHANES. Our modeled obesity prevalence aligned closely with measured data from five validation states, with a 0.64 percentage point mean difference (range: 0.23-1.39) and a high correlation coefficient (r=0.96, P=0.009). Estimated state-specific childhood obesity prevalence ranged from 11.0 to 20.4%. CONCLUSION: Uncorrected estimates of childhood obesity prevalence from NSCH vary widely from measured national data, from a 278% overestimate among males aged 2-5 years to a 44% underestimate among females aged 14-17 years. This study demonstrates the validity of the CHOICES matching methods to correct the bias of parent-reported BMI data and highlights the need for public release of more recent data from the 2011 to 2012 NSCH.


Subject(s)
Pediatric Obesity/epidemiology , Public Health Surveillance , Public Health , Self Report/standards , Adolescent , Body Mass Index , Child , Child, Preschool , Female , Humans , Male , Nutrition Surveys , Parents , Pediatric Obesity/prevention & control , Policy Making , Prevalence , United States/epidemiology
3.
Phys Rev Lett ; 112(16): 167201, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24815664

ABSTRACT

In the majority of magnetic systems the surface is required to order at the same temperature as the bulk. In the present Letter, we report a distinct and unexpected surface magnetic phase transition at a lower temperature than the Néel temperature. Employing grazing incidence x-ray resonant magnetic scattering, we have observed the near-surface behavior of uranium dioxide. UO2 is a noncollinear, triple-q, antiferromagnet with the U ions on a face-centered cubic lattice. Theoretical investigations establish that at the surface the energy increase-due to the lost bonds-is reduced when the spins near the surface rotate, gradually losing their component normal to the surface. At the surface the lowest-energy spin configuration has a double-q (planar) structure. With increasing temperature, thermal fluctuations saturate the in-plane crystal field anisotropy at the surface, leading to soft excitations that have ferromagnetic XY character and are decoupled from the bulk. The structure factor of a finite two-dimensional XY model fits the experimental data well for several orders of magnitude of the scattered intensity. Our results support a distinct magnetic transition at the surface in the Kosterlitz-Thouless universality class.

4.
J Thromb Haemost ; 3(3): 541-51, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15748245

ABSTRACT

Megakaryocytes were analyzed for their ability to endocytose factor V to define the cellular mechanisms regulating this process. In contrast to fibrinogen, factor V was endocytosed by megakaryocytes derived from CD34(+) cells or megakaryocyte-like cell lines, but not by platelets. CD41(+)ex vivo-derived megakaryocytes endocytosed factor V, as did subpopulations of the megakaryocyte-like cells MEG-01, and CMK. Similar observations were made for fibrinogen. Phorbol diester-induced megakaryocytic differentiation of the cell lines resulted in a substantial increase in endocytosis of both proteins as compared to untreated cells that did not merely reflect their disparate plasma concentrations. Factor IX, which does not associate with platelets or megakaryocytes, was not endocytosed by any of the cells examined. Endocytosis of factor V by megakaryocytes proceeds through a specific and independent mechanism as CHRF-288 cells endocytosed fibrinogen but not factor V, and the presence of other plasma proteins had no effect on the endocytosis of factor V by MEG-01 cells. Furthermore, as the endocytosis of factor V was also demonstrated to occur through a clathrin-dependent mechanism, these combined data demonstrate that endocytosis of factor V by megakaryocytes occurs via a specific, independent, and most probably receptor-mediated, event.


Subject(s)
Clathrin/physiology , Endocytosis , Factor V/metabolism , Megakaryocytes/physiology , Cell Differentiation , Cell Lineage , Cell Membrane/metabolism , Cell Membrane/physiology , Cells, Cultured , Humans , Megakaryocytes/cytology , Megakaryocytes/ultrastructure , Protein Binding
5.
Rapid Commun Mass Spectrom ; 15(18): 1649-61, 2001.
Article in English | MEDLINE | ID: mdl-11555863

ABSTRACT

A liquid-phase three-dimensional protein separation method has been developed that is used to separate the cytosolic fraction of a HEL cell lysate via isoelectric focusing (IEF), nonporous silica (NPS) reversed-phase high-performance liquid chromatography (RP-HPLC) and electrospray ionization time-of-flight mass spectrometry (ESI-TOFMS), respectively. Several hundred unique protein molecular weights were observed in a pI range from 4.8 to 8.5 and a mass range from 5 to 85 kDa. Proteins were positively identified by analysis of the pI (+/-0.5 pI units), an intact protein molecular weight (+/-150 ppm), and peptide mass mapping results. Using the molecular weight (MW) and peptide mapping results of identified proteins it was possible to characterize their posttranslational (PTMs) and/or sequence modifications. PTMs were detected on both forms of cytosolic actin, heat shock 90 beta, HINT and alpha-enolase. Sequence modifications or conflicts were observed for beta-and gamma-actin, ATP beta-synthase and heat shock 90 beta. IEF-NPS-RP-HPLC/ESI-TOFMS was used to determine experimental pI, MW and relative hydrophobicity values for each protein detected. This data was used to generate a 2-D pI-MS protein map, where proteins are displayed according to their pI and molecular weight. Protein molecular weight peaks are represented as bands in the 2-D pI-MS image where the gray scale of each band is proportional to the intensity of the protein molecular weight peak. In addition, a third hydrophobicity dimension (%B) was added as the % acetonitrile elution to generate a 3-D pI-MS-%B plot where each protein can be tagged according to three parameters.


Subject(s)
Leukemia, Erythroblastic, Acute/metabolism , Neoplasm Proteins/chemistry , Amino Acid Sequence , Cell Line , Chromatography, High Pressure Liquid , Cytosol/chemistry , Electrophoresis , Humans , Hydrolysis , Isoelectric Focusing , Molecular Sequence Data , Molecular Weight , Neoplasm Proteins/isolation & purification , Peptide Mapping , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypsin , Tumor Cells, Cultured
6.
Cancer Res ; 61(17): 6487-93, 2001 Sep 01.
Article in English | MEDLINE | ID: mdl-11522645

ABSTRACT

We have shown previously that mitotic spindle inhibitors allow the c-Myconcoprotein to uncouple mitosis from DNA synthesis, resulting in the acquisition of tetraploidy. This can also occur in the absence of spindle inhibition if c-Myc deregulation is combined with inactivation of the p53 tumor suppressor. Under these conditions, cyclin B1 protein is induced but retains its normal cell cycle regulation. We now show that the cyclin B1 promoter is directly but oppositely regulated by c-Myc and p53. Enforced expression of cyclin B1 also induces tetraploidy, either after mitotic spindle inhibition or in the absence of such inhibition if cyclin B1 is coexpressed with c-Myc. Cyclin B1 represents a new class of c-Myc target genes that is also regulated by p53. It is also the first identified downstream effector of c-Myc able to produce the chromosomal instability that characterizes virtually all tumor cells.


Subject(s)
Cyclin B/genetics , Gene Expression Regulation/physiology , Ploidies , Proto-Oncogene Proteins c-myc/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Line , Cyclin B/biosynthesis , Cyclin B1 , Humans , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Tumor Suppressor Protein p53/biosynthesis
8.
Blood Cells Mol Dis ; 27(3): 677-90, 2001.
Article in English | MEDLINE | ID: mdl-11482883

ABSTRACT

This paper addresses some of the important aspects of stem cell commitment to the bone cell lineage examining the various types of precursor cells, their responses to cytokines and other extracellular influences, and recent observations on the biochemical and molecular control of lineage-specific gene expression. The process of osteopoiesis involves the proliferation and maturation of primitive precursor cells into functional osteoblasts. The bone cells purportedly originate from mesenchymal stem cells that commit to the osteogenic cell lineage becoming osteoprogenitor cells, preosteoblasts, osteoblasts, and osteocytes. Further understanding of this developmental process requires that lineage-specific markers be identified for the various populations of bone cells and their precursors, that cell separation techniques be established so that cells of the osteogenic lineage can be purified at different stages of differentiation, and that these isolated cells are studied under serum-free, chemically defined conditions.


Subject(s)
Bone Marrow Cells/cytology , Cell Lineage , Osteogenesis/physiology , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Differentiation , Cell Division/drug effects , Growth Substances/pharmacology , Growth Substances/physiology , Humans , Immunophenotyping , Stem Cells/cytology
9.
Nat Biotechnol ; 18(9): 954-8, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10973215

ABSTRACT

Tissue engineering of human bone is a complex process, as the functional development of bone cells requires that regulatory signals be temporally and spatially ordered. The role of three-dimensional cellular interactions is well understood in embryonic osteogenesis, but in vitro correlates are lacking. Here we report that in vitro serum-free transforming growth factor (TGF)-beta1 stimulation of osteogenic cells immediately after passage results in the formation of three-dimensional cellular condensations (bone cell spheroids) within 24 to 48 hours. In turn, bone cell spheroid formation results in the up-regulation of several bone-related proteins (e.g., alkaline phosphatase, type I collagen, osteonectin) during days 3-7, and the concomitant formation of micro-crystalline bone. This system of ex vivo bone formation should provide important information on the physiological, biological and molecular basis of osteogenesis.


Subject(s)
Biomedical Engineering/methods , Bone Transplantation , Bone and Bones/cytology , Bone and Bones/physiology , Alkaline Phosphatase/metabolism , Biotechnology , Blotting, Western , Bone Development , Bone Marrow/metabolism , Bone and Bones/ultrastructure , Cell Division , Cell Line , Cells, Cultured , Collagen/metabolism , Culture Media, Serum-Free/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Integrins/metabolism , Microscopy, Electron , Osteogenesis/physiology , Osteonectin/metabolism , Regeneration/physiology , Spectroscopy, Fourier Transform Infrared , Spectrum Analysis, Raman , Time Factors , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/therapeutic use , Transforming Growth Factor beta1 , Up-Regulation
10.
Exp Hematol ; 28(7): 815-25, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10907643

ABSTRACT

OBJECTIVE: Much remains to be learned about the intimate relationship between bone marrow and its surrounding tissue: the bone. We hypothesized that bone marrow accessory cell populations might regulate the development of human bone precursor cells. MATERIALS AND METHODS: We used immunologic phenotyping, and isolation methods to fractionate subpopulations of nonadherent, low-density (NALD) human bone marrow cells. These cells were examined for their ability to support the serum-free survival, proliferation, and expression of bone proteins by highly purified populations of human bone precursor cells. Quantitative assessment of the accessory cell populations as well as human bone precursor cells phenotype was performed using multiparameter flow cytometry. Bone protein expression was evaluated by immunocytochemistry, Western analysis, and enzymatic analysis (for alkaline phosphatase activity). RESULTS: Human bone marrow contains a cell population that stimulates the development of purified bone precursor cells. Feeder-layer studies demonstrate that these osteopoietic accessory cells (OACs) do not require cell-cell interaction to promote bone precursor cell development but, rather, produce soluble molecules responsible for their effects. Flow cytometric analyses reveal that bone marrow derived B cells, T cells, macrophages, natural killer cells, and endothelial cells do not produce this stimulatory factor. The (growth) factor cannot be replaced by addition of exogenous cytokines. The isolation of human transforming growth factor beta receptor type II (TGF-betaRII)-positive cells increases OAC-specific activity in bone cell ex vivo expansion cultures. Moreover, isolation of OAC bone marrow cells characterized by high TGF-betaRII expression, relatively low cellular complexity, and small size yields a population that is highly enriched for OACs. CONCLUSION: We conclude that human bone marrow contains a population of OACs that are an obligate requirement for the early phases of bone cell development ex vivo.


Subject(s)
Antigen-Presenting Cells/physiology , Bone Marrow Cells/physiology , Osteoblasts/physiology , Cell Adhesion , Cell Separation , Cells, Cultured , Cytokines/biosynthesis , Flow Cytometry , Humans , Immunophenotyping , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
11.
Crit Rev Eukaryot Gene Expr ; 10(3-4): 259-71, 2000.
Article in English | MEDLINE | ID: mdl-11272468

ABSTRACT

An understanding of the disorders of bone formation clearly requires insights into the complex regulatory events occurring during the evolution of bone precursor cells into osteoblasts. Moreover, a rational approach to therapeutic interventions that might alter the clinical course of bone disorders must take into consideration the exact nature of the developmental control mechanism(s) being affected during the disease process. The process of osteopoiesis involves the proliferation and maturation of primitive precursor cells into functional osteoblasts. The bone cell lineage originates from mesenchymal stem cells that commit to the osteogenic cell lineage becoming osteoprogenitor cells, preosteoblasts, osteoblasts, and osteocytes. In order to understand how different regulatory signals coordinate bone cell development, it is important to study the responses of bone progenitor cells to different microenviromental signals. This requires that lineage markers be identified for the various populations of bone cells and their precursors, that cell separation techniques be established so that cells of the osteogenic lineage can be purified at different stages of differentiation, and that these isolated cells are studied under serum-free, chemically defined conditions. This review focuses on the current understanding of bone progenitor cell development, examining the various types of precursor cells, their responses to cytokines and other extracellular influences, and recent observations on the biochemical and molecular control of lineage-specific gene expression. Although the emphasis is on human cells, the importance of work using rodent cells goes without saying, and is addressed where relevant.


Subject(s)
Bone Development , Bone and Bones/cytology , Stem Cells/cytology , Humans
12.
Br J Haematol ; 107(4): 895-902, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10606901

ABSTRACT

To address trafficking of transplanted marrow cells immediately after intravenous infusion, we examined the early fate of infused non-adherent, low-density donor bone marrow cells in a syngeneic mouse model. The presence of infused donor cells, marked with indium-111 oxine (111In), with the fluorescent dye PKH26, or by a detectable transgene marker, was evaluated at 3-48 h in a variety of tissues, including peripheral blood. All three cell-marking methods indicated a rapid (< 4 h) influx of cells into the bone marrow, liver, spleen, muscle and other tissues. Moreover, these tissues remained positive for the 48 h observation period. Interestingly, analysis of PKH26-positive cells in non-myeloablated animals demonstrated that approximately 17% of infused donor marrow cells localized to the marrow space within 15 h, whereas a smaller proportion of donor cells (approximately 1-2%) localized to the marrow in recipients preconditioned by irradiation. In an effort to enrich for cells that specifically home to the bone marrow, PKH26-labelled donor marrow cells were recovered from the first host and infused into a secondary recipient. Although this was a phenotypically undefined population of cells, no increase was observed in the relative fraction of PKH26-labelled cells returning or 'homing' to the marrow of the second recipient. Taken together, these data suggest both that marrow engraftment may be mediated by non-specific 'seeding' rather than a specific homing signal, and that efficient targeting of transplanted cells to the marrow is a complex multifaceted process.


Subject(s)
Bone Marrow Transplantation/physiology , Hematopoiesis/physiology , Animals , Cell Movement/physiology , Flow Cytometry , Infusions, Intravenous , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction/methods
13.
Curr Opin Hematol ; 6(3): 159-63, 1999 May.
Article in English | MEDLINE | ID: mdl-10226736

ABSTRACT

The recent cloning of the thrombopoietin gene, and the production of recombinant protein, have allowed studies on both its biological actions and clinical utility. Thrombopoietin not only affects the cells of the megakaryocytic lineage, but has a diverse set of cellular targets. In particular, it stimulates the ex vivo expansion of hematopoietic stem/progenitor cells suggesting that it may play a role in transplantation studies. Pre-clinical but limited clinical studies indicate that under defined conditions, thrombopoietin may accelerate white blood cell count and platelet recoveries following myelosuppression or radiotherapy.


Subject(s)
Hematopoietic Stem Cells/cytology , Thrombopoietin/physiology , DNA/metabolism , Humans , Thrombopoietin/genetics
14.
J Gerontol A Biol Sci Med Sci ; 54(2): B54-62, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10051851

ABSTRACT

The ability to purify and characterize phenotypic markers of human bone precursor cells provides an important means to study the basis of age- or disease-related changes in osteogenesis. Utilizing immunologically purified and characterized populations of human bone preosteoblast-like cells, we demonstrate that distinct age-related alterations occur in bone cell phenotypic markers, and additionally document the presence of a subpopulation of elderly individuals who express markedly reduced amounts of bone proteins. These findings provide insights into the early phases of bone cell development, and provide a means for evaluating age- and/or disease-mediated changes in bone cell development.


Subject(s)
Aging/pathology , Bone Marrow Cells/cytology , Phenotype , Stem Cells/cytology , Adolescent , Adult , Aged , Aged, 80 and over , Aging/genetics , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Morphogenetic Proteins/pharmacology , Cell Differentiation/drug effects , Cell Lineage , Child , Child, Preschool , Disease , Female , Genetic Markers/genetics , Humans , Infant , Male , Middle Aged , Osteoblasts/cytology , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Osteonectin/genetics , Osteonectin/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Transforming Growth Factor beta/pharmacology
15.
Oncogene ; 18(5): 1177-84, 1999 Feb 04.
Article in English | MEDLINE | ID: mdl-10022123

ABSTRACT

p53 monitors genomic integrity at the G1 and G2/M cell cycle checkpoints. Cells lacking p53 may show gene amplification as well as the polyploidy or aneuploidy typical of many tumors. The pathways through which this develops, however, are not well defined. We demonstrate here that the combination of p53 inactivation and c-myc overexpression in diploid cells markedly accelerates the spontaneous development of tetraploidy. This is not seen with either N-myc or L-myc. Tetraploidy is accompanied by significantly higher levels of cyclin B and its associated cdc2 kinase activity. Mitotic spindle poisons accelerate the appearance of tetraploidy in cells either lacking functional p53 or overexpressing c-myc whereas the combination is additive. Restoration of p53 function in cells overexpressing c-myc causing rapid apoptosis, indicating that cells yet to become tetraploid have nonetheless suffered irreversible genomic and/or mitotic spindle damage. In the face of normal p53 function, such damage would either be repaired or trigger apoptotis. We propose that loss of p53 and overexpression of c-myc permits the emergence and survival of cells with increasingly severe damage and the eventual development of tetraploidy.


Subject(s)
Polyploidy , Proto-Oncogene Proteins c-myc/biosynthesis , Repressor Proteins , Tumor Suppressor Protein p53/deficiency , Animals , CDC2 Protein Kinase/metabolism , Cell Line , Cyclin B/metabolism , Diploidy , Genetic Vectors , Mice , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Proto-Oncogene Proteins c-myc/genetics , Recombinant Proteins/biosynthesis , Spindle Apparatus/pathology
16.
Cell Growth Differ ; 9(8): 639-50, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9716181

ABSTRACT

Despite a growing understanding of the biochemical mechanisms controlling the cell cycle, information regarding the temporal ordering of S phase and M phase remains scarce. Polyploid cells represent a useful model for examining S- and M-phase control, because their cell cycle machinery must be modulated to retain high levels of DNA content (ploidy) within a single nucleus. To evaluate the mechanisms of S-phase control during the process of polyploidization, we investigated the modulations that occur in cyclin-dependent kinase (CDK) complexes during the induction of megakaryocyte differentiation in human erythroleukemia cells. We report that during polyploidization, megakaryocytic human erythroleukemia cells undergo a dramatic modulation in the subunit composition of G1-associated and S phase-associated CDK complexes and a marked increase in their specific activities. This, in turn, is facilitated by a differential loss of the p21 or p27 CDK-inhibitory protein/kinase-inhibitory proteins (CIP/KIP) bound to specific cyclin/CDK complexes. The data show that the loss of S- and M-phase control in polyploid cells occurs within the context of an up-regulated function in those CDK complexes associated with both G1-S-phase transit and S-phase progression. Additional studies regarding the regulation of these complex CDK interactions will be important to understand cell cycle control in such diverse processes as megakaryocyte differentiation or the types of genomic instability that occur in cancer cells.


Subject(s)
CDC2-CDC28 Kinases , Cell Cycle Proteins , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , G1 Phase , Polyploidy , Protein Serine-Threonine Kinases/metabolism , S Phase , Tumor Suppressor Proteins , Cell Differentiation , Cell Nucleus/metabolism , Cyclin A/genetics , Cyclin A/metabolism , Cyclin D3 , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase Inhibitor p21 , Cyclin-Dependent Kinase Inhibitor p27 , Cyclins/genetics , Cyclins/physiology , Half-Life , Histones/metabolism , Humans , Leukemia, Erythroblastic, Acute , Megakaryocytes , Microtubule-Associated Proteins/metabolism , Mitosis , Phosphorylation , Tumor Cells, Cultured
17.
Semin Hematol ; 35(3): 192-9, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9685165

ABSTRACT

The events underlying the commitment and differentiation of megakaryocytes are poorly understood, particularly with respect to understanding the biochemical and molecular mechanisms regulating this process. These regulatory events begin with the interaction of multiple microenvironmental signals (eg, cytokines, extracellular matrix) with specific cell surface receptors, extend through a signal transduction cascade, and end with transcriptional activation of megakaryocyte-specific genes. This article focuses on the cellular, biochemical, and molecular control of megakaryocyte differentiation events, whereas data on the ligands, receptors, and signal transduction are found elsewhere in this issue. The first area discussed is the classification of functional categories of the cells of the megakaryocyte lineage: identifying those cells which respond to proliferative signals, those which mark the transition from the proliferating cell compartment to mature cells, and the mature, post-mitotic platelet-shedding cells. The transitional cells, the pro-megakaryoblasts, are covered in some detail as these cells are physiologically important both in their early response to thrombopoietic stress, and for their unique capacity to continue to synthesize DNA during their differentiation. Finally, recent data on the control of the process of megakaryocyte polyploidization, as well as the molecular control of megakaryocyte commitment are discussed.


Subject(s)
Cell Differentiation , Megakaryocytes/cytology , Animals , Cell Differentiation/genetics , DNA/metabolism , Hematopoiesis , Hematopoietic Stem Cells/cytology , Humans
18.
Blood ; 91(11): 4118-26, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9596657

ABSTRACT

Little is known concerning the interaction of thrombopoietin (TPO) with other megakaryocyte-active cytokines in directing the early events of megakaryocyte development. Culture of CD34(+) cells in interleukins (IL) -1, -6, -11, plus stem cell factor (SCF; S) results in a 10- to 12-fold expansion in total cell numbers, whereas total CD41(+) megakaryocytes are expanded approximately 120-fold over input levels. Addition of TPO to IL-1, -6, -11, S generates a biphasic proliferation of CD41(+) cells, accelerates their rate of production, and results in an ex vivo expansion of more than 200-fold. The addition of Flt-3 ligand (FL) increases CD41+ cell expansion to approximately 380-fold over input levels. In the absence of TPO, approximately 95% of the expanded cells show the phenotype of promegakaryoblasts; TPO and/or FL addition increases CD41 antigen density and ploidy in a subpopulation of promegakaryoblasts. A moderate (approximately sevenfold) expansion of megakaryocyte progenitor cells (colony-forming unit-megakaryocyte) occurs in the presence of IL-1, -6, -11, S, and the addition of TPO to this cocktail yields an approximately 17-fold expansion. We conclude that early proliferative events in megakaryocyte development in vitro are regulated by multiple cytokines, and that TPO markedly affects these early developmental steps. However, by itself, TPO is neither necessary nor sufficient to generate a full proliferative/maturational in vitro response within the megakaryocyte compartment. TPO clearly affects terminal differentiation and the development of (some) high-ploidy human megakaryocytes. However, its limited in vitro actions on human cell polyploidization suggest that additional megakaryocyte-active cytokines or other signals are essential for the maximal development of human megakaryocytes.


Subject(s)
Cytokines/pharmacology , Hematopoietic Stem Cells/cytology , Megakaryocytes/cytology , Thrombopoietin/physiology , Adult , Antigens, CD34/metabolism , Cell Differentiation/drug effects , Culture Media, Serum-Free , Hematopoiesis , Hematopoietic Stem Cells/drug effects , Humans , Interleukin-1/pharmacology , Interleukin-11/pharmacology , Interleukin-6/pharmacology , Membrane Proteins/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Stem Cell Factor/pharmacology
19.
Nat Biotechnol ; 15(10): 1007-11, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9335055

ABSTRACT

The ability to isolate functional populations of hematopoietic progenitor cells is important to the process of hematopoietic cell transplantation and to the understanding of hematopoietic cell biology in health and disease. We show that a subpopulation of human bone marrow hematopoietic cells bearing the pan-hematopoietic antigen CD34 also binds galactose-conjugated proteins. This lectin-positive sub-population represents approximately 0.1 to 0.5% of the total bone marrow cells, and contains 100% of the hematopoietic progenitor cells. The galactose-binding lectin on these cells is specific for this sugar. Additionally, highly proliferative hematopoietic progenitor cells with very primitive phenotypes, including a newly identified progenitor cell that produces multiple lineages, express this lectin.


Subject(s)
Antigens, CD34/analysis , Cell Separation/methods , Hemagglutinins/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Antigens, CD34/genetics , Cell Adhesion , Cells, Cultured , Flow Cytometry/methods , Fluorescein-5-isothiocyanate/analogs & derivatives , Galactose/chemistry , Galactosides/chemistry , Galectins , Hemagglutinins/chemistry , Hemagglutinins/genetics , Humans , Serum Albumin, Bovine/chemistry
20.
Mol Biol Cell ; 7(2): 209-23, 1996 Feb.
Article in English | MEDLINE | ID: mdl-8688553

ABSTRACT

The pathways that regulate the S-phase events associated with the control of DNA replication are poorly understood. The bone marrow megakaryocytes are unique in that they leave the diploid (2C) state to differentiate, synthesizing 4 to 64 times the normal DNA content within a single nucleus, a process known as endomitosis. Human erythroleukemia (HEL) cells model this process, becoming polyploid during phorbol diester-induced megakaryocyte differentiation. The mitotic arrest occurring in these polyploid cells involves novel alterations in the cdk1/cyclin B1 complex: a marked reduction in cdk1 protein levels, and an elevated and sustained expression of cyclin B1. Endomitotic cells thus lack cdk1/cyclin B1-associated H1-histone kinase activity. Constitutive over-expression of cdk1 in endomitotic cells failed to re-initiate normal mitotic events even though cdk1 was present in a 10-fold excess. This was due to an inability of cyclin-B1 to physically associate with cdk1. Nonetheless, endomitotic cyclin B1 possesses immunoprecipitable H1-histone kinase activity, and specifically translocates to the nucleus. We conclude that mitosis is abrogated during endomitosis due to the absence of cdk1 and the failure to form M-phase promoting factor, resulting in a disassociation of mitosis from the completion of S-phase. Further studies on cyclin and its interacting proteins should be informative in understanding endomitosis and cell cycle control.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin B , Cyclins/metabolism , Polyploidy , Cell Cycle , Cell Division , Cell Nucleus/metabolism , Cyclin B1 , Humans , Mitosis , Protamine Kinase/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...