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










Publication year range
1.
Nucleic Acids Res ; 51(5): 2464-2484, 2023 03 21.
Article in English | MEDLINE | ID: mdl-36762498

ABSTRACT

Riboswitches regulate downstream gene expression by binding cellular metabolites. Regulation of translation initiation by riboswitches is posited to occur by metabolite-mediated sequestration of the Shine-Dalgarno sequence (SDS), causing bypass by the ribosome. Recently, we solved a co-crystal structure of a prequeuosine1-sensing riboswitch from Carnobacterium antarcticum that binds two metabolites in a single pocket. The structure revealed that the second nucleotide within the gene-regulatory SDS, G34, engages in a crystal contact, obscuring the molecular basis of gene regulation. Here, we report a co-crystal structure wherein C10 pairs with G34. However, molecular dynamics simulations reveal quick dissolution of the pair, which fails to reform. Functional and chemical probing assays inside live bacterial cells corroborate the dispensability of the C10-G34 pair in gene regulation, leading to the hypothesis that the compact pseudoknot fold is sufficient for translation attenuation. Remarkably, the C. antarcticum aptamer retained significant gene-regulatory activity when uncoupled from the SDS using unstructured spacers up to 10 nucleotides away from the riboswitch-akin to steric-blocking employed by sRNAs. Accordingly, our work reveals that the RNA fold regulates translation without SDS sequestration, expanding known riboswitch-mediated gene-regulatory mechanisms. The results infer that riboswitches exist wherein the SDS is not embedded inside a stable fold.


Subject(s)
Protein Biosynthesis , Riboswitch , Binding Sites , Gene Expression Regulation , Molecular Dynamics Simulation , Nucleic Acid Conformation , Ribosomes/genetics , Ribosomes/metabolism
2.
Blood Adv ; 6(10): 3072-3089, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35139174

ABSTRACT

Primitive erythropoiesis is a critical component of the fetal cardiovascular network and is essential for the growth and survival of the mammalian embryo. The need to rapidly establish a functional cardiovascular system is met, in part, by the intravascular circulation of primitive erythroid precursors that mature as a single semisynchronous cohort. To better understand the processes that regulate erythroid precursor maturation, we analyzed the proteome, metabolome, and lipidome of primitive erythroblasts isolated from embryonic day (E) 10.5 and E12.5 of mouse gestation, representing their transition from basophilic erythroblast to orthochromatic erythroblast (OrthoE) stages of maturation. Previous transcriptional and biomechanical characterizations of these precursors have highlighted a transition toward the expression of protein elements characteristic of mature red blood cell structure and function. Our analysis confirmed a loss of organelle-specific protein components involved in messenger RNA processing, proteostasis, and metabolism. In parallel, we observed metabolic rewiring toward the pentose phosphate pathway, glycolysis, and the Rapoport-Luebering shunt. Activation of the pentose phosphate pathway in particular may have stemmed from increased expression of hemoglobin chains and band 3, which together control oxygen-dependent metabolic modulation. Increased expression of several antioxidant enzymes also indicated modification to redox homeostasis. In addition, accumulation of oxylipins and cholesteryl esters in primitive OrthoE cells was paralleled by increased transcript levels of the p53-regulated cholesterol transporter (ABCA1) and decreased transcript levels of cholesterol synthetic enzymes. The present study characterizes the extensive metabolic rewiring that occurs in primary embryonic erythroid precursors as they prepare to enucleate and continue circulating without internal organelles.


Subject(s)
Erythroblasts , Proteomics , Animals , Embryo, Mammalian/metabolism , Erythroblasts/metabolism , Erythropoiesis/genetics , Hemoglobins/metabolism , Mammals , Mice
3.
Epigenetics Chromatin ; 13(1): 16, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32178723

ABSTRACT

BACKGROUND: SETD8 is the sole methyltransferase capable of mono-methylating histone H4, lysine 20. SETD8 and H4K20me1 play a role in a number of essential biologic processes, including cell cycle progression, establishment of higher order chromatin structure, and transcriptional regulation. SETD8 is highly expressed in erythroid cells and erythroid deletion of Setd8 is embryonic lethal by embryonic day 11.5 (E11.5) due to profound anemia, suggesting that it has an erythroid-specific function. The function of SETD8 in the hemopoietic system is poorly understood. The goal of our study was to gain insights into the function of SETD8 during erythroid differentiation. RESULTS: We performed ATAC-seq (assay for transposase-accessible chromatin) on sorted populations of E10.5 Setd8 mutant and control erythroblasts. Accessibility profiles were integrated with expression changes and a mark of heterochromatin (H3K27me3) performed in wild-type E10.5 erythroblasts to further understand the role of SETD8 in erythropoiesis. Data integration identified regions of greater chromatin accessibility in Setd8 mutant cells that co-located with H3K27me3 in wild-type E10.5 erythroblasts suggesting that these regions, and their associated genes, are repressed during normal erythropoiesis. The majority of these more accessible regions were located in promoters and they frequently co-located with the NFY complex. Pathway analysis of genes identified through data integration revealed stemness-related pathways. Among those genes were multiple transcriptional regulators active in multipotent progenitors, but repressed during erythroid differentiation including Hhex, Hlx, and Gata2. Consistent with a role for SETD8 in erythroid specification, SETD8 expression is up-regulated upon erythroid commitment, and Setd8 disruption impairs erythroid colony forming ability. CONCLUSION: Taken together, our results suggest that SETD8 is an important regulator of the chromatin landscape during erythroid differentiation, particularly at promoters. Our results also identify a novel role for Setd8 in the establishment of appropriate patterns of lineage-restricted gene expression during erythroid differentiation.


Subject(s)
Chromatin Assembly and Disassembly , Erythropoiesis , Histone-Lysine N-Methyltransferase/metabolism , Transcription Factors/genetics , Animals , Cell Line , Cells, Cultured , Erythroblasts/cytology , Erythroblasts/metabolism , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , Transcription Factors/metabolism
4.
J Biomol Tech ; 31(2): 47-56, 2020 07.
Article in English | MEDLINE | ID: mdl-31966025

ABSTRACT

Small RNAs (smRNAs) are important regulators of many biologic processes and are now most frequently characterized using Illumina sequencing. However, although standard RNA sequencing library preparation has become routine in most sequencing facilities, smRNA sequencing library preparation has historically been challenging because of high input requirements, laborious protocols involving gel purifications, inability to automate, and a lack of benchmarking standards. Additionally, studies have suggested that many of these methods are nonlinear and do not accurately reflect the amounts of smRNAs in vivo. Recently, a number of new kits have become available that permit lower input amounts and less laborious, gel-free protocol options. Several of these new kits claim to reduce RNA ligase-dependent sequence bias through novel adapter modifications and to lessen adapter-dimer contamination in the resulting libraries. With the increasing number of smRNA kits available, understanding the relative strengths of each method is crucial for appropriate experimental design. In this study, we systematically compared 9 commercially available smRNA library preparation kits as well as NanoString probe hybridization across multiple study sites. Although several of the new methodologies do reduce the amount of artificially over- and underrepresented microRNAs (miRNAs), we observed that none of the methods was able to remove all of the bias in the library preparation. Identical samples prepared with different methods show highly varied levels of different miRNAs. Even so, many methods excelled in ease of use, lower input requirement, fraction of usable reads, and reproducibility across sites. These differences may help users select the most appropriate methods for their specific question of interest.


Subject(s)
Gene Library , High-Throughput Nucleotide Sequencing/standards , MicroRNAs/genetics , Sequence Analysis, RNA/standards , MicroRNAs/isolation & purification , Reproducibility of Results , Software
5.
Sci Rep ; 7(1): 14381, 2017 10 30.
Article in English | MEDLINE | ID: mdl-29084993

ABSTRACT

Erythropoietin (EPO) and its receptor are highly expressed in the developing nervous system, and exogenous EPO therapy is potentially neuroprotective, however the epigenetic and transcriptional changes downstream of EPO signaling in neural cells are not well understood. To delineate epigenetic changes associated with EPO signaling, we compared histone H3 lysine 4 dimethylation (H3K4me2) in EPO treated and control fetal neural progenitor cells, identifying 1,150 differentially bound regions. These regions were highly enriched near protein coding genes and had significant overlap with H4Acetylation, a mark of active regulatory elements. Motif analyses and co-occupancy studies revealed a complex regulatory network underlying the differentially bound regions, including previously identified mediators of EPO signaling (STAT5, STAT3), and novel factors such as REST, an epigenetic modifier central to neural differentiation and plasticity, and NRF1, a key regulator of antioxidant response and mitochondrial biogenesis. Global transcriptome analyses on neural tubes isolated from E9.0 EpoR-null and littermate control embryos validated our in vitro findings, further suggesting a role for REST and NRF1 downstream of EPO signaling. These data support a role for EPO in regulating the survival, proliferation, and differentiation of neural progenitor cells, and suggest a basis for its function in neural development and neuroprotection.


Subject(s)
Erythropoietin/metabolism , Neural Stem Cells/metabolism , Cell Differentiation/physiology , Cells, Cultured , DNA-Binding Proteins/genetics , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Erythropoietin/physiology , Fetal Stem Cells/metabolism , Fetus/physiology , Gene Regulatory Networks , Humans , Neural Stem Cells/physiology , Neurons/metabolism , Nuclear Respiratory Factor 1/drug effects , Receptors, Erythropoietin/metabolism , Repressor Proteins/drug effects , Signal Transduction/physiology , Trans-Activators/metabolism , Transcriptome/genetics
6.
Cell Rep ; 21(9): 2376-2383, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29186677

ABSTRACT

Erythropoiesis is a highly regulated process that generates enucleate red blood cells from committed erythroid progenitors. Chromatin condensation culminating in enucleation is a defining feature of this process. Setd8 is the sole enzyme that can mono-methylate histone H4, lysine 20 and is highly expressed in erythroblasts compared to most other cell types. Erythroid Setd8 deletion results in embryonic lethality from severe anemia due to impaired erythroblast survival and proliferation. Setd8 protein levels are also uniquely regulated in erythroblasts, suggesting a cell-type-specific role for Setd8 during terminal maturation. Consistent with this hypothesis, Setd8 Δ/Δ erythroblasts have profound defects in transcriptional repression, chromatin condensation, and heterochromatin accumulation. Together, these results suggest that Setd8, used by most cells to promote mitotic chromatin condensation, is an essential aspect of the transcriptional repression and chromatin condensation that are hallmarks of terminal erythroid maturation.


Subject(s)
Erythroblasts/enzymology , Histone-Lysine N-Methyltransferase/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Erythroblasts/metabolism , Erythropoiesis/genetics , Erythropoiesis/physiology , Female , Heterochromatin/genetics , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/metabolism , Mice , Pregnancy
7.
Mol Cell Biol ; 35(12): 2059-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25848090

ABSTRACT

Setd8 is the sole histone methyltransferase in mammals capable of monomethylating histone H4 lysine 20 (H4K20me1). Setd8 is expressed at significantly higher levels in erythroid cells than any other cell or tissue type, suggesting that Setd8 has an erythroid-cell-specific function. To test this hypothesis, stable Setd8 knockdown was established in extensively self-renewing erythroblasts (ESREs), a well-characterized, nontransformed model of erythroid maturation. Knockdown of Setd8 resulted in impaired erythroid maturation characterized by a delay in hemoglobin accumulation, larger mean cell area, persistent ckit expression, incomplete nuclear condensation, and lower rates of enucleation. Setd8 knockdown did not alter ESRE proliferation or viability or result in accumulation of DNA damage. Global gene expression analyses following Setd8 knockdown demonstrated that in erythroid cells, Setd8 functions primarily as a repressor. Most notably, Gata2 expression was significantly higher in knockdown cells than in control cells and Gata2 knockdown rescued some of the maturation impairments associated with Setd8 disruption. Setd8 occupies critical regulatory elements in the Gata2 locus, and knockdown of Setd8 resulted in loss of H4K20me1 and gain of H4 acetylation at the Gata2 1S promoter. These results suggest that Setd8 is an important regulator of erythroid maturation that works in part through repression of Gata2 expression.


Subject(s)
Erythroid Cells/cytology , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Histone-Lysine N-Methyltransferase/metabolism , Acetylation , Animals , Cells, Cultured , Erythroblasts/cytology , Erythroblasts/metabolism , Erythroid Cells/metabolism , Erythropoiesis , GATA2 Transcription Factor/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/genetics
8.
Haematologica ; 98(11): 1778-87, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23894012

ABSTRACT

Primitive erythroid cells, the first red blood cells produced in the mammalian embryo, are necessary for embryonic survival. Erythropoietin and its receptor EpoR, are absolutely required for survival of late-stage definitive erythroid progenitors in the fetal liver and adult bone marrow. Epo- and Epor-null mice die at E13.5 with a lack of definitive erythrocytes. However, the persistence of circulating primitive erythroblasts raises questions about the role of erythropoietin/EpoR in primitive erythropoiesis. Using Epor-null mice and a novel primitive erythroid 2-step culture we found that erythropoietin is not necessary for specification of primitive erythroid progenitors. However, Epor-null embryos develop a progressive, profound anemia by E12.5 as primitive erythroblasts mature as a synchronous cohort. This anemia results from reduced primitive erythroblast proliferation associated with increased p27 expression, from advanced cellular maturation, and from markedly elevated rates of apoptosis associated with an imbalance in pro- and anti-apoptotic gene expression. Both mouse and human primitive erythroblasts cultured without erythropoietin also undergo accelerated maturation and apoptosis at later stages of maturation. We conclude that erythropoietin plays an evolutionarily conserved role in promoting the proliferation, survival, and appropriate timing of terminal maturation of primitive erythroid precursors.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Erythroblasts/physiology , Erythropoietin/physiology , Animals , Cell Lineage/physiology , Cell Survival/physiology , Cells, Cultured , Humans , Mice , Mice, Transgenic
9.
BMC Syst Biol ; 7: 38, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23675896

ABSTRACT

BACKGROUND: Hematopoietic ontogeny is characterized by overlapping waves of primitive, fetal definitive, and adult definitive erythroid lineages. Our aim is to identify differences in the transcriptional control of these distinct erythroid cell maturation pathways by inferring and analyzing gene-interaction networks from lineage-specific expression datasets. Inferred networks are strongly connected and do not fit a scale-free model, making it difficult to identify essential regulators using the hub-essentiality standard. RESULTS: We employed a semi-supervised machine learning approach to integrate measures of network topology with expression data to score gene essentiality. The algorithm was trained and tested on the adult and fetal definitive erythroid lineages. When applied to the primitive erythroid lineage, 144 high scoring transcription factors were found to be differentially expressed between the primitive and adult definitive erythroid lineages, including all expressed STAT-family members. Differential responses of primitive and definitive erythroblasts to a Stat3 inhibitor and IFNγ in vitro supported the results of the computational analysis. Further investigation of the original expression data revealed a striking signature of Stat1-related genes in the adult definitive erythroid network. Among the potential pathways known to utilize Stat1, interferon (IFN) signaling-related genes were expressed almost exclusively within the adult definitive erythroid network. CONCLUSIONS: In vitro results support the computational prediction that differential regulation and downstream effectors of STAT signaling are key factors that distinguish the transcriptional control of primitive and definitive erythroid cell maturation.


Subject(s)
Artificial Intelligence , Computational Biology/methods , Erythropoiesis/genetics , Gene Expression Regulation , Gene Regulatory Networks , Interferons/genetics , STAT Transcription Factors/genetics , Adult , Algorithms , Animals , Humans , Mice , Signal Transduction/genetics
10.
Blood ; 121(6): e5-e13, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23243273

ABSTRACT

Erythroid ontogeny is characterized by overlapping waves of primitive and definitive erythroid lineages that share many morphologic features during terminal maturation but have marked differences in cell size and globin expression. In the present study, we compared global gene expression in primitive, fetal definitive, and adult definitive erythroid cells at morphologically equivalent stages of maturation purified from embryonic, fetal, and adult mice. Surprisingly, most transcriptional complexity in erythroid precursors is already present by the proerythroblast stage. Transcript levels are markedly modulated during terminal erythroid maturation, but housekeeping genes are not preferentially lost. Although primitive and definitive erythroid lineages share a large set of nonhousekeeping genes, annotation of lineage-restricted genes shows that alternate gene usage occurs within shared functional categories, as exemplified by the selective expression of aquaporins 3 and 8 in primitive erythroblasts and aquaporins 1 and 9 in adult definitive erythroblasts. Consistent with the known functions of Aqp3 and Aqp8 as H2O2 transporters, primitive, but not definitive, erythroblasts preferentially accumulate reactive oxygen species after exogenous H2O2 exposure. We have created a user-friendly Web site (http://www.cbil.upenn.edu/ErythronDB) to make these global expression data readily accessible and amenable to complex search strategies by the scientific community.


Subject(s)
Erythroid Cells/metabolism , Erythropoiesis/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Animals , Aquaporin 1/genetics , Aquaporin 3/genetics , Aquaporins/genetics , Cell Lineage/genetics , Cells, Cultured , Erythroblasts/metabolism , Erythrocytes/metabolism , Female , Hematopoietic System/cytology , Hematopoietic System/embryology , Hematopoietic System/growth & development , Mice , Mice, Inbred ICR , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
11.
Clin Lymphoma Myeloma Leuk ; 11(3): 289-92, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21658658

ABSTRACT

Lymphoma is an increasingly recognized complication of tumor necrosis factor (TNF) inhibition for the treatment of autoimmune and inflammatory disease; the majority of these cases are non-Hodgkin lymphomas (NHL). The impact of withdrawing TNF inhibition therapy in cases of lymphoma is not well described. A woman with Crohn's disease (CD) was diagnosed with Hodgkin lymphoma (HL) and subsequently went into remission with standard chemotherapy. Her CD later worsened, requiring initiation of adalimumab, a TNF inhibitor. Ten months later, she was found to have recurrence of HL. When she opted against additional treatment for the lymphoma, the TNF inhibitor was discontinued. Three months later, the measurable sites of disease had completely regressed. It can be concluded that HL is a potential complication of treatment with TNF inhibitors. Withdrawal of immunosuppression may be a consideration for patients treated for lymphoproliferative disorders including HL. Maintenance of an intact immune system may be important for prevention of lymphoma relapse. Further understanding of this complex interaction will help clinicians determine in which patients these agents have a favorable risk-benefit ratio.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Crohn Disease/drug therapy , Hodgkin Disease/pathology , Neoplasm Regression, Spontaneous/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adalimumab , Anti-Inflammatory Agents/adverse effects , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Cholecystitis/complications , Cholecystitis/diagnostic imaging , Cholecystitis/surgery , Fatal Outcome , Female , Hematoma/etiology , Hodgkin Disease/chemically induced , Hodgkin Disease/diagnostic imaging , Humans , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Lymphatic Diseases/diagnostic imaging , Lymphatic Diseases/pathology , Middle Aged , Radiography , Retroperitoneal Space , Ultrasonography
12.
Am J Surg Pathol ; 35(2): 177-89, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21263238

ABSTRACT

The normal counterparts of mantle cell lymphoma (MCL) are naive, quiescent B cells that have not been processed through the germinal center (GC). For this reason, although lymphomas arising from GC or post-GC B cells often exhibit plasmacytic differentiation, MCL rarely presents with plasmacytic features. Seven cases of MCL with a monotypic plasma cell (PC) population were collected from 6 centers and were studied by immunohistochemistry, fluorescence immunophenotyping and interphase cytogenetics as a tool for the investigation of neoplasms analysis, capillary gel electrophoresis, and restriction fragment length polymorphism of immunoglobulin heavy chain analysis of microdissections of each of the MCL and PC populations to assess their clonal relationship. The clinical presentation was rather unusual compared with typical MCL, with 2 cases arising from the extranodal soft tissues of the head. All MCL cases were morphologically and immunohistochemically typical, bearing the t(11;14)(q13;q32). In all cases, the PC population was clonal. In 5 of the 7 cases, the MCL and PC clones showed identical restriction fragments, indicating a common clonal origin of the neoplastic population. The 2 cases with clonal diversity denoted the coexistence of 2 different tumors in a composite lymphoma/PC neoplasm. Our findings suggest that MCL can present with a PC component that is often clonally related to the lymphoma, representing a rare but unique biological variant of this tumor.


Subject(s)
Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Lymphoma, Mantle-Cell/genetics , Plasma Cells/pathology , Translocation, Genetic/genetics , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Clone Cells , DNA, Neoplasm/analysis , Female , Humans , Immunoenzyme Techniques , Immunoglobulin Heavy Chains/genetics , Immunophenotyping , In Situ Hybridization, Fluorescence , Lymphoid Tissue/metabolism , Lymphoid Tissue/pathology , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/pathology , Male , Microdissection , Middle Aged , Plasma Cells/metabolism , Polymorphism, Restriction Fragment Length
13.
Int J Dev Biol ; 54(6-7): 1011-8, 2010.
Article in English | MEDLINE | ID: mdl-20711979

ABSTRACT

Erythropoiesis in adult mammals is characterized by the progressive maturation of hematopoietic stem cells to lineage-specific progenitors, to morphologically identifiable precursors which enucleate to form mature erythrocytes. In contrast, primitive erythropoiesis is characterized by the appearance within the yolk sac of a transient, lineage-restricted progenitor population which generates a wave of erythroid precursors. These precursors undergo progressive maturation in the bloodstream, characterized by nuclear condensation and embryonic hemoglobin accumulation. This process is dependent on erythropoietin signaling through its cognate receptor, as well as the function of several erythroid-specific transcription factors, including GATA1 and EKLF. Targeted disruption of genes in the mouse that result in failure of the emergence or maturation of the primitive erythroid lineage leads to early fetal death, indicating that the primitive erythroid lineage is necessary for survival of the mammalian embryo. While it was thought for over a century that primitive erythroid cells were uniquely nucleated mammalian red cells, it is now recognized that they, like their definitive erythroid counterparts, enucleate to form reticulocytes and pyrenocytes. This surprising finding indicates that the primitive erythroid lineage is indeed mammalian, rather than non-mammalian, in character.


Subject(s)
Embryo, Mammalian/metabolism , Erythroid Cells/metabolism , Erythropoiesis/genetics , Animals , Embryo, Mammalian/blood supply , Embryo, Mammalian/cytology , Erythroid Cells/cytology , GATA1 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Humans , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout
14.
Blood ; 107(4): 1665-72, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16263786

ABSTRACT

Mammals have 2 distinct erythroid lineages. The primitive erythroid lineage originates in the yolk sac and generates a cohort of large erythroblasts that terminally differentiate in the bloodstream. The definitive erythroid lineage generates smaller enucleated erythrocytes that become the predominant cell in fetal and postnatal circulation. These lineages also have distinct globin expression patterns. Our studies in primary murine primitive erythroid cells indicate that betaH1 is the predominant beta-globin transcript in the early yolk sac. Thus, unlike the human, murine beta-globin genes are not up-regulated in the order of their chromosomal arrangement. As primitive erythroblasts mature from proerythroblasts to reticulocytes, they undergo a betaH1- to epsilony-globin switch, up-regulate adult beta1- and beta2-globins, and down-regulate zeta-globin. These changes in transcript levels correlate with changes in RNA polymerase II density at their promoters and transcribed regions. Furthermore, the epsilony- and betaH1-globin genes in primitive erythroblasts reside within a single large hyperacetylated domain. These data suggest that this "maturational" betaH1- to epsilony-globin switch is dynamically regulated at the transcriptional level. Globin switching during ontogeny is due not only to the sequential appearance of primitive and definitive lineages but also to changes in globin expression as primitive erythroblasts mature in the bloodstream.


Subject(s)
Genes, Switch , Globins/genetics , Animals , Cell Differentiation , Embryo, Mammalian , Erythroblasts/cytology , Erythroblasts/physiology , Erythrocytes/cytology , Erythrocytes/physiology , Histone Acetyltransferases/metabolism , Histones/metabolism , Mice , RNA Polymerase II/metabolism
15.
Blood ; 104(1): 19-25, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15031208

ABSTRACT

The enucleated definitive erythrocytes of mammals are unique in the animal kingdom. The observation that yolk sac-derived primitive erythroid cells in mammals circulate as nucleated cells has led to the conjecture that they are related to the red cells of fish, amphibians, and birds that remain nucleated throughout their life span. In mice, primitive red cells express both embryonic and adult hemoglobins, whereas definitive erythroblasts accumulate only adult hemoglobins. We investigated the terminal differentiation of murine primitive red cells with use of antibodies raised to embryonic beta H1-globin. Primitive erythroblasts progressively enucleate between embryonic days 12.5 and 16.5, generating mature primitive erythrocytes that are similar in size to their nucleated counterparts. These enucleated primitive erythrocytes circulate as late as 5 days after birth. The enucleation of primitive red cells in the mouse embryo has not previously been well recognized because it coincides with the emergence of exponentially expanding numbers of definitive erythrocytes from the fetal liver. Our studies establish a new paradigm in the understanding of primitive erythropoiesis and support the concept that primitive erythropoiesis in mice shares many similarities with definitive erythropoiesis of mammals.


Subject(s)
Erythroblasts/physiology , Mice/embryology , Yolk Sac/physiology , Animals , Cell Count/methods , Cell Differentiation , Cell Size , Embryo, Mammalian/blood supply , Erythroblasts/cytology , Erythroblasts/ultrastructure , Erythrocytes/cytology , Erythropoiesis/physiology , Female , Gene Expression , Globins/biosynthesis , Globins/chemistry , Globins/immunology , Immunohistochemistry , Microscopy, Phase-Contrast , Pregnancy , Yolk Sac/cytology
16.
Blood ; 101(5): 1669-76, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12406884

ABSTRACT

To better understand the relationship between the embryonic hematopoietic and vascular systems, we investigated the establishment of circulation in mouse embryos by examining the redistribution of yolk sac-derived primitive erythroblasts and definitive hematopoietic progenitors. Our studies revealed that small numbers of erythroblasts first enter the embryo proper at 4 to 8 somite pairs (sp) (embryonic day 8.25 [E8.25]), concomitant with the proposed onset of cardiac function. Hours later (E8.5), most red cells remained in the yolk sac. Although the number of red cells expanded rapidly in the embryo proper, a steady state of approximately 40% red cells was not reached until 26 to 30 sp (E10). Additionally, erythroblasts were unevenly distributed within the embryo's vasculature before 35 sp. These data suggest that fully functional circulation is established after E10. This timing correlated with vascular remodeling, suggesting that vessel arborization, smooth muscle recruitment, or both are required. We also examined the distribution of committed hematopoietic progenitors during early embryogenesis. Before E8.0, all progenitors were found in the yolk sac. When normalized to circulating erythroblasts, there was a significant enrichment (20- to 5-fold) of progenitors in the yolk sac compared with the embryo proper from E9.5 to E10.5. These results indicated that the yolk sac vascular network remains a site of progenitor production and preferential adhesion even as the fetal liver becomes a hematopoietic organ. We conclude that a functional vascular system develops gradually and that specialized vascular-hematopoietic environments exist after circulation becomes fully established.


Subject(s)
Blood Circulation , Embryo, Mammalian/blood supply , Animals , Blood Cell Count , Blood Vessels/embryology , Embryonic and Fetal Development , Erythroblasts/cytology , Erythrocyte Count , Gestational Age , Hematopoietic Stem Cells/cytology , Liver/embryology , Mice , Mice, Inbred ICR , Neovascularization, Physiologic , Yolk Sac/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...