Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 162
Filter
1.
iScience ; 25(1): 103680, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35036870

ABSTRACT

lncRAP2 is a conserved cytoplasmic lncRNA enriched in adipose tissue and required for adipogenesis. Using purification and in vivo interactome analyses, we show that lncRAP2 forms complexes with proteins that stabilize mRNAs and modulate translation, among them Igf2bp2. Surveying transcriptome-wide Igf2bp2 client mRNAs in white adipocytes reveals selective binding to mRNAs encoding adipogenic regulators and energy expenditure effectors, including adiponectin. These same target proteins are downregulated when either Igf2bp2 or lncRAP2 is downregulated, hindering adipocyte lipolysis. Proteomics and ribosome profiling show this occurs predominantly through mRNA accumulation, as lncRAP2-Igf2bp2 complex binding does not impact translation efficiency. Phenome-wide association studies reveal specific associations of genetic variants within both lncRAP2 and Igf2bp2 with body mass and type 2 diabetes, and both lncRAP2 and Igf2bp2 are suppressed in adipose depots of obese and diabetic individuals. Thus, the lncRAP2-Igf2bp2 complex potentiates adipose development and energy expenditure and is associated with susceptibility to obesity-linked diabetes.

2.
PLoS One ; 16(11): e0259353, 2021.
Article in English | MEDLINE | ID: mdl-34731223

ABSTRACT

Low plasma levels of Proprotein Convertase Subtilisin/Kexin 9 (PCSK9) are associated with decreased low-density lipoprotein (LDL) cholesterol and a reduced risk of cardiovascular disease. PCSK9 binds to the epidermal growth factor-like repeat A (EGFA) domain of LDL receptors (LDLR), very low-density lipoprotein receptors (VLDLR), apolipoprotein E receptor 2 (ApoER2), and lipoprotein receptor-related protein 1 (LRP1) and accelerates their degradation, thus acting as a key regulator of lipid metabolism. Antibody and RNAi-based PCSK9 inhibitor treatments lower cholesterol and prevent cardiovascular incidents in patients, but their high-cost hampers market penetration. We sought to develop a safe, long-term and one-time solution to treat hyperlipidemia. We created a cDNA encoding a chimeric protein in which the extracellular N- terminus of red blood cells (RBCs) specific glycophorin A was fused to the LDLR EGFA domain and introduced this gene into mouse bone marrow hematopoietic stem and progenitor cells (HSPCs). Following transplantation into irradiated mice, the animals produced RBCs with the EGFA domain (EGFA-GPA RBCs) displayed on their surface. These animals showed significantly reduced plasma PCSK9 (66.5% decrease) and reduced LDL levels (40% decrease) for as long as 12 months post-transplantation. Furthermore, the EGFA- GPA mice remained lean for life and maintained normal body weight under a high-fat diet. Hematopoietic stem cell gene therapy can generate red blood cells expressing an EGFA-glycophorin A chimeric protein as a practical and long-term strategy for treating chronic hyperlipidemia and obesity.


Subject(s)
Cholesterol, LDL/blood , Down-Regulation , Glycophorins/genetics , Hyperlipidemias/prevention & control , Proprotein Convertase 9/blood , Receptors, LDL/genetics , Animals , Body Weight , Cells, Cultured , Diet, High-Fat/adverse effects , Erythrocytes/metabolism , Female , Genetic Engineering , Glycophorins/chemistry , HEK293 Cells , Humans , Hyperlipidemias/chemically induced , Hyperlipidemias/metabolism , Mice , Pregnancy , Receptors, LDL/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cell Transplantation , Transduction, Genetic
3.
Exp Hematol ; 89: 1-12, 2020 09.
Article in English | MEDLINE | ID: mdl-32798645

ABSTRACT

I am deeply honored to receive the International Society for Experimental Hematology (ISEH) 2020 Donald Metcalf Lecture Award. Although I am not a physician and have had no formal training in hematology, I have had the privilege of working with some of the top hematologists in the world, beginning in 1970 when Dr. David Nathan was a sabbatical visitor in my laboratory and introduced me to hematological diseases.  And I take this award to be given not just to me but to an exceptional group of MD and PhD trainees and visitors in my laboratory who have cloned and characterized many proteins and RNAs important for red cell development and function. Many of these projects involved taking exceptionally large risks in developing and employing novel experimental technologies. Unsurprisingly, all of these trainees have gone on to become leaders in hematology and, more broadly, in molecular cell biology and molecular medicine. To illustrate some of the challenges we have faced and the technologies we had to develop, I have chosen several of our multiyear projects to describe in some detail: elucidating the regulation of translation of α- and ß-globin mRNAs and the defect in beta thalassemia in the 1970s; cloning the Epo receptor and several red cell membrane proteins in the 1980s and 1990s; and more recently, determining the function of many microRNAs and long noncoding RNAs in red cell development. I summarize how we are currently utilizing single-cell transcriptomics (scRNAseq) to understand how dividing transit-amplifying burst-forming unit erythroid progenitors balance the need for more progenitor cells with the need for terminally differentiated erythroid cells, and to identify drugs potentially useful in treating Epo-resistant anemias such as Diamond Blackfan anemia. I hope that the lessons I learned in managing these diverse fellows and projects, initially without having grants to support them, will be helpful to others who would like to undertake ambitious and important lines of research in hematology.


Subject(s)
Erythroid Precursor Cells/metabolism , Hematology/history , Molecular Biology/history , Receptors, Erythropoietin/history , beta-Thalassemia/genetics , Cloning, Molecular , Erythrocytes/metabolism , Erythrocytes/pathology , Erythroid Precursor Cells/cytology , Erythropoiesis/genetics , Gene Expression , History, 20th Century , History, 21st Century , Humans , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , alpha-Globins/genetics , alpha-Globins/metabolism , beta-Globins/genetics , beta-Globins/metabolism , beta-Thalassemia/metabolism , beta-Thalassemia/pathology
4.
Proc Natl Acad Sci U S A ; 117(26): 15055-15065, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32554489

ABSTRACT

Phosphocholine phosphatase-1 (PHOSPHO1) is a phosphocholine phosphatase that catalyzes the hydrolysis of phosphocholine (PC) to choline. Here we demonstrate that the PHOSPHO1 transcript is highly enriched in mature brown adipose tissue (BAT) and is further induced by cold and isoproterenol treatments of BAT and primary brown adipocytes. In defining the functional relevance of PHOPSPHO1 in BAT thermogenesis and energy metabolism, we show that PHOSPHO1 knockout mice are cold-tolerant, with higher expression of thermogenic genes in BAT, and are protected from high-fat diet-induced obesity and development of insulin resistance. Treatment of mice with the PHOSPHO1 substrate phosphocholine is sufficient to induce cold tolerance, thermogenic gene expression, and allied metabolic benefits. Our results reveal a role of PHOSPHO1 as a negative regulator of BAT thermogenesis, and inhibition of PHOSPHO1 or enhancement of phosphocholine represent innovative approaches to manage the metabolic syndrome.


Subject(s)
Adipose Tissue, Brown/physiology , Phosphoric Monoester Hydrolases/genetics , Phosphorylcholine/metabolism , Thermogenesis , Adipocytes, Brown/enzymology , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/enzymology , Animals , Cold Temperature , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoric Monoester Hydrolases/deficiency
5.
Dev Cell ; 49(1): 118-129.e7, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30827895

ABSTRACT

The nature of cell-state transitions during the transit-amplifying phases of many developmental processes-hematopoiesis in particular-is unclear. Here, we use single-cell RNA sequencing to demonstrate a continuum of transcriptomic states in committed transit-amplifying erythropoietic progenitors, which correlates with a continuum of proliferative potentials in these cells. We show that glucocorticoids enhance erythrocyte production by slowing the rate of progression through this developmental continuum of transit-amplifying progenitors, permitting more cell divisions prior to terminal erythroid differentiation. Mechanistically, glucocorticoids prolong expression of genes that antagonize and slow induction of genes that drive terminal erythroid differentiation. Erythroid progenitor daughter cell pairs have similar transcriptomes with or without glucocorticoid stimulation, indicating largely symmetric cell division. Thus, the rate of progression along a developmental continuum dictates the absolute number of erythroid cells generated from each transit-amplifying progenitor, suggesting a paradigm for regulating the total output of differentiated cells in numerous other developmental processes.


Subject(s)
Blood Cells/metabolism , Cell Proliferation/genetics , Erythroid Precursor Cells/metabolism , Hematopoiesis/genetics , Animals , Blood Cells/cytology , Cell Differentiation/genetics , Cell Division/genetics , Cells, Cultured , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , Erythroid Precursor Cells/cytology , Erythropoiesis/genetics , Glucocorticoids/genetics , High-Throughput Nucleotide Sequencing/methods , Mice , Single-Cell Analysis/methods , Transcriptome/genetics
6.
J Biol Chem ; 293(51): 19797-19811, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30366982

ABSTRACT

Erythropoietin (EPO) signaling is critical to many processes essential to terminal erythropoiesis. Despite the centrality of iron metabolism to erythropoiesis, the mechanisms by which EPO regulates iron status are not well-understood. To this end, here we profiled gene expression in EPO-treated 32D pro-B cells and developing fetal liver erythroid cells to identify additional iron regulatory genes. We determined that FAM210B, a mitochondrial inner-membrane protein, is essential for hemoglobinization, proliferation, and enucleation during terminal erythroid maturation. Fam210b deficiency led to defects in mitochondrial iron uptake, heme synthesis, and iron-sulfur cluster formation. These defects were corrected with a lipid-soluble, small-molecule iron transporter, hinokitiol, in Fam210b-deficient murine erythroid cells and zebrafish morphants. Genetic complementation experiments revealed that FAM210B is not a mitochondrial iron transporter but is required for adequate mitochondrial iron import to sustain heme synthesis and iron-sulfur cluster formation during erythroid differentiation. FAM210B was also required for maximal ferrochelatase activity in differentiating erythroid cells. We propose that FAM210B functions as an adaptor protein that facilitates the formation of an oligomeric mitochondrial iron transport complex, required for the increase in iron acquisition for heme synthesis during terminal erythropoiesis. Collectively, our results reveal a critical mechanism by which EPO signaling regulates terminal erythropoiesis and iron metabolism.


Subject(s)
Erythroid Cells/metabolism , Erythropoietin/metabolism , Ferrochelatase/metabolism , Heme/biosynthesis , Iron/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Erythroid Cells/cytology , Erythropoiesis , HEK293 Cells , Humans , Membrane Proteins/chemistry , Mice , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/chemistry , Protein Transport
7.
Hematol Oncol Clin North Am ; 32(4): 701-712, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30047421

ABSTRACT

Diamond-Blackfan anemia (DBA) is a severe congenital hypoplastic anemia caused by mutation in a ribosomal protein gene. Major clinical issues concern the optimal management of patients resistant to steroids, the first-line therapy. Hematopoietic stem cell transplantation is indicated in young patients with an HLA-matched unaffected sibling donor, and recent results with matched unrelated donor transplants indicate that these patients also do well. When neither steroids nor a transplant is possible red cell transfusions are required, and iron loading is rapid in some DBA patients, so effective chelation is vital. Also discussed are novel treatments under investigation for DBA.


Subject(s)
Anemia, Diamond-Blackfan , Erythrocyte Transfusion , Hematopoietic Stem Cell Transplantation , Mutation , Tissue Donors , Allografts , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/metabolism , Anemia, Diamond-Blackfan/pathology , Anemia, Diamond-Blackfan/therapy , Humans , Siblings
8.
Nat Commun ; 8(1): 2115, 2017 12 13.
Article in English | MEDLINE | ID: mdl-29235464

ABSTRACT

Brown adipose tissue (BAT) metabolism influences glucose homeostasis and metabolic health in mice and humans. Sympathetic stimulation of ß-adrenergic receptors in response to cold induces proliferation, differentiation, and UCP1 expression in pre-adipocytes and mature brown adipocytes. Here we show that spleen tyrosine kinase (SYK) is upregulated during brown adipocyte differentiation and activated by ß-adrenergic stimulation. Deletion or inhibition of SYK, a kinase known for its essential roles in the immune system, blocks brown and white pre-adipocyte proliferation and differentiation in vitro, and results in diminished expression of Ucp1 and other genes regulating brown adipocyte function in response to ß-adrenergic stimulation. Adipocyte-specific SYK deletion in mice reduces BAT mass and BAT that developed consisted of SYK-expressing brown adipocytes that had escaped homozygous Syk deletion. SYK inhibition in vivo represses ß-agonist-induced thermogenesis and oxygen consumption. These results establish SYK as an essential mediator of brown fat formation and function.


Subject(s)
Adipocytes, Brown/enzymology , Adipose Tissue, Brown/metabolism , Cell Differentiation , Syk Kinase/metabolism , Adipocytes, Brown/cytology , Animals , Cell Proliferation , Cells, Cultured , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Syk Kinase/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
9.
Mol Biol Cell ; 28(22): 2908-2910, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29084906

ABSTRACT

Advancement of science depends on thoughtfully mentoring a rare group of scientists that are highly educated, creative, and motivated-and that come from every country in the world. On the basis of my own experiences, I suggest ways to recruit top young scientists of both genders, support their development into leading researchers, and advise them about careers inside and outside of academia. Creating a family-friendly environment within the laboratory and the institution is crucial to these efforts.


Subject(s)
Mentoring/trends , Personnel Selection/methods , Humans , Laboratories , Mentors , Research Personnel
10.
Nat Commun ; 8(1): 423, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28871080

ABSTRACT

A short half-life in the circulation limits the application of therapeutics such as single-domain antibodies (VHHs). We utilize red blood cells to prolong the circulatory half-life of VHHs. Here we present VHHs against botulinum neurotoxin A (BoNT/A) on the surface of red blood cells by expressing chimeric proteins of VHHs with Glycophorin A or Kell. Mice whose red blood cells carry the chimeric proteins exhibit resistance to 10,000 times the lethal dose (LD50) of BoNT/A, and transfusion of these red blood cells into naive mice affords protection for up to 28 days. We further utilize an improved CD34+ culture system to engineer human red blood cells that express these chimeric proteins. Mice transfused with these red blood cells are resistant to highly lethal doses of BoNT/A. We demonstrate that engineered red blood cells expressing VHHs can provide prolonged prophylactic protection against bacterial toxins without inducing inhibitory immune responses and illustrates the potentially broad translatability of our strategy for therapeutic applications.The therapeutic use of single-chain antibodies (VHHs) is limited by their short half-life in the circulation. Here the authors engineer mouse and human red blood cells to express VHHs against botulinum neurotoxin A (BoNT/A) on their surface and show that an infusion of these cells into mice confers long lasting protection against a high dose of BoNT/A.


Subject(s)
Botulinum Toxins, Type A/toxicity , Erythrocytes/metabolism , Genetic Engineering , Single-Domain Antibodies/genetics , Animals , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/metabolism , Botulinum Toxins, Type A/metabolism , Botulism/etiology , Botulism/therapy , Erythrocyte Transfusion , Erythrocytes/virology , Erythroid Precursor Cells/metabolism , Erythroid Precursor Cells/transplantation , Erythroid Precursor Cells/virology , Genetic Vectors/genetics , Genetic Vectors/metabolism , Glycophorins/genetics , Glycophorins/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mice , Mice, Inbred C57BL , Retroviridae/genetics , Retroviridae/metabolism , Single-Domain Antibodies/administration & dosage , Single-Domain Antibodies/metabolism
11.
Proc Natl Acad Sci U S A ; 114(38): 10107-10112, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28864529

ABSTRACT

An effect of thyroid hormone (TH) on erythropoiesis has been known for more than a century but the molecular mechanism(s) by which TH affects red cell formation is still elusive. Here we demonstrate an essential role of TH during terminal human erythroid cell differentiation; specific depletion of TH from the culture medium completely blocked terminal erythroid differentiation and enucleation. Treatment with TRß agonists stimulated premature erythroblast differentiation in vivo and alleviated anemic symptoms in a chronic anemia mouse model by regulating erythroid gene expression. To identify factors that cooperate with TRß during human erythroid terminal differentiation, we conducted RNA-seq in human reticulocytes and identified nuclear receptor coactivator 4 (NCOA4) as a critical regulator of terminal differentiation. Furthermore, Ncoa4-/- mice are anemic in perinatal periods and fail to respond to TH by enhanced erythropoiesis. Genome-wide analysis suggests that TH promotes NCOA4 recruitment to chromatin regions that are in proximity to Pol II and are highly associated with transcripts abundant during terminal differentiation. Collectively, our results reveal the molecular mechanism by which TH functions during red blood cell formation, results that are potentially useful to treat certain anemias.


Subject(s)
Cell Differentiation , Nuclear Receptor Coactivators/metabolism , Reticulocytes/metabolism , Thyroid Hormone Receptors beta/metabolism , Thyroid Hormones/metabolism , Animals , Chromatin/genetics , Chromatin/metabolism , Genome-Wide Association Study , Humans , Mice , Mice, Knockout , Nuclear Receptor Coactivators/genetics , Thyroid Hormone Receptors beta/genetics , Thyroid Hormones/genetics
12.
Blood ; 130(18): 1965-1975, 2017 11 02.
Article in English | MEDLINE | ID: mdl-28928124

ABSTRACT

Long noncoding RNAs (lncRNAs) are increasingly recognized as vital components of gene programs controlling cell differentiation and function. Central to their functions is an ability to act as scaffolds or as decoys that recruit or sequester effector proteins from their DNA, RNA, or protein targets. lncRNA-modulated effectors include regulators of transcription, chromatin organization, RNA processing, and translation, such that lncRNAs can influence gene expression at multiple levels. Here we review the current understanding of how lncRNAs help coordinate gene expression to modulate cell fate in the hematopoietic system. We focus on a growing number of mechanistic studies to synthesize emerging principles of lncRNA function, emphasizing how they facilitate diversification of gene programming during development. We also survey how disrupted lncRNA function can contribute to malignant transformation, highlighting opportunities for therapeutic intervention in specific myeloid and lymphoid cancers. Finally, we discuss challenges and prospects for further elucidation of lncRNA mechanisms.


Subject(s)
Hematologic Neoplasms/genetics , Hematopoiesis/genetics , RNA, Long Noncoding/genetics , Animals , Embryonic Development/genetics , Humans , Models, Biological , Proteins/metabolism , RNA, Long Noncoding/metabolism
13.
Cell Rep ; 19(12): 2503-2514, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28636939

ABSTRACT

Enhancer-derived RNAs are thought to act locally by contributing to their parent enhancer function. Whether large domains of clustered enhancers (super-enhancers) also produce cis-acting RNAs, however, remains unclear. Unlike typical enhancers, super-enhancers form large spans of robustly transcribed chromatin, amassing capped and polyadenylated RNAs that are sufficiently abundant to sustain trans functions. Here, we show that one such RNA, alncRNA-EC7/Bloodlinc, is transcribed from a super-enhancer of the erythroid membrane transporter SLC4A1/BAND3 but diffuses beyond this site. Bloodlinc localizes to trans-chromosomal loci encoding critical regulators and effectors of terminal erythropoiesis and directly binds chromatin-organizing and transcription factors, including the chromatin attachment factor HNRNPU. Inhibiting Bloodlinc or Hnrnpu compromises the terminal erythropoiesis gene program, blocking red cell production, whereas expressing Bloodlinc ectopically stimulates this program and can promote erythroblast proliferation and enucleation in the absence of differentiation stimuli. Thus, Bloodlinc is a trans-acting super-enhancer RNA that potentiates red blood cell development.


Subject(s)
Erythrocytes/physiology , Erythropoiesis , RNA, Long Noncoding/physiology , Animals , Cells, Cultured , Enhancer Elements, Genetic , Erythroid Cells/metabolism , Gene Expression Regulation , Humans , Mice , Transcription, Genetic
14.
Elife ; 62017 05 29.
Article in English | MEDLINE | ID: mdl-28553927

ABSTRACT

Heme is required for survival of all cells, and in most eukaryotes, is produced through a series of eight enzymatic reactions. Although heme production is critical for many cellular processes, how it is coupled to cellular differentiation is unknown. Here, using zebrafish, murine, and human models, we show that erythropoietin (EPO) signaling, together with the GATA1 transcriptional target, AKAP10, regulates heme biosynthesis during erythropoiesis at the outer mitochondrial membrane. This integrated pathway culminates with the direct phosphorylation of the crucial heme biosynthetic enzyme, ferrochelatase (FECH) by protein kinase A (PKA). Biochemical, pharmacological, and genetic inhibition of this signaling pathway result in a block in hemoglobin production and concomitant intracellular accumulation of protoporphyrin intermediates. Broadly, our results implicate aberrant PKA signaling in the pathogenesis of hematologic diseases. We propose a unifying model in which the erythroid transcriptional program works in concert with post-translational mechanisms to regulate heme metabolism during normal development.


Subject(s)
A Kinase Anchor Proteins/metabolism , Erythropoietin/metabolism , GATA1 Transcription Factor/metabolism , Heme/biosynthesis , Signal Transduction , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Humans , Mice , Mitochondrial Membranes/metabolism , Zebrafish
15.
Proc Natl Acad Sci U S A ; 114(12): 3157-3162, 2017 03 21.
Article in English | MEDLINE | ID: mdl-28270614

ABSTRACT

Current therapies for autoimmune diseases rely on traditional immunosuppressive medications that expose patients to an increased risk of opportunistic infections and other complications. Immunoregulatory interventions that act prophylactically or therapeutically to induce antigen-specific tolerance might overcome these obstacles. Here we use the transpeptidase sortase to covalently attach disease-associated autoantigens to genetically engineered and to unmodified red blood cells as a means of inducing antigen-specific tolerance. This approach blunts the contribution to immunity of major subsets of immune effector cells (B cells, CD4+ and CD8+ T cells) in an antigen-specific manner. Transfusion of red blood cells expressing self-antigen epitopes can alleviate and even prevent signs of disease in experimental autoimmune encephalomyelitis, as well as maintain normoglycemia in a mouse model of type 1 diabetes.

16.
Sci Transl Med ; 9(376)2017 02 08.
Article in English | MEDLINE | ID: mdl-28179501

ABSTRACT

Diamond-Blackfan anemia (DBA) is a congenital disorder characterized by the failure of erythroid progenitor differentiation, severely curtailing red blood cell production. Because many DBA patients fail to respond to corticosteroid therapy, there is considerable need for therapeutics for this disorder. Identifying therapeutics for DBA requires circumventing the paucity of primary patient blood stem and progenitor cells. To this end, we adopted a reprogramming strategy to generate expandable hematopoietic progenitor cells from induced pluripotent stem cells (iPSCs) from DBA patients. Reprogrammed DBA progenitors recapitulate defects in erythroid differentiation, which were rescued by gene complementation. Unbiased chemical screens identified SMER28, a small-molecule inducer of autophagy, which enhanced erythropoiesis in a range of in vitro and in vivo models of DBA. SMER28 acted through autophagy factor ATG5 to stimulate erythropoiesis and up-regulate expression of globin genes. These findings present an unbiased drug screen for hematological disease using iPSCs and identify autophagy as a therapeutic pathway in DBA.


Subject(s)
Anemia, Diamond-Blackfan/drug therapy , Drug Discovery , Hematopoietic Stem Cells/metabolism , Allyl Compounds/pharmacology , Anemia, Diamond-Blackfan/pathology , Antigens, CD34/metabolism , Autophagy/drug effects , Autophagy-Related Protein 5/metabolism , Cell Differentiation/drug effects , Cellular Reprogramming , Erythroid Cells/drug effects , Erythroid Cells/pathology , Erythropoiesis/drug effects , Genetic Complementation Test , Globins/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Quinazolines/pharmacology
17.
Blood ; 128(23): 2637-2641, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27777239

ABSTRACT

Burst-forming unit erythroid progenitors (BFU-Es) are so named based on their ability to generate in methylcellulose culture large colonies of erythroid cells that consist of "bursts" of smaller erythroid colonies derived from the later colony-forming unit erythroid progenitor erythropoietin (Epo)-dependent progenitors. "Early" BFU-E cells forming large BFU-E colonies presumably have higher capacities for self-renewal than do "late" BFU-Es forming small colonies, but the mechanism underlying this heterogeneity remains unknown. We show that the type III transforming growth factor ß (TGF-ß) receptor (TßRIII) is a marker that distinguishes early and late BFU-Es. Transient elevation of TßRIII expression promotes TGF-ß signaling during the early BFU-E to late BFU-E transition. Blocking TGF-ß signaling using a receptor kinase inhibitor increases early BFU-E cell self-renewal and total erythroblast production, suggesting the usefulness of this type of drug in treating Epo-unresponsive anemias.


Subject(s)
Antigens, Differentiation/metabolism , Erythrocytes/metabolism , Erythroid Precursor Cells/metabolism , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Anemia/metabolism , Anemia/therapy , Animals , Erythrocytes/cytology , Erythroid Precursor Cells/cytology , Erythropoietin/metabolism , Humans , Mice
18.
Cell ; 165(7): 1672-1685, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27315481

ABSTRACT

Long intergenic noncoding RNAs (lincRNAs) are important regulators of gene expression. Although lincRNAs are expressed in immune cells, their functions in immunity are largely unexplored. Here, we identify an immunoregulatory lincRNA, lincRNA-EPS, that is precisely regulated in macrophages to control the expression of immune response genes (IRGs). Transcriptome analysis of macrophages from lincRNA-EPS-deficient mice, combined with gain-of-function and rescue experiments, revealed a specific role for this lincRNA in restraining IRG expression. Consistently, lincRNA-EPS-deficient mice manifest enhanced inflammation and lethality following endotoxin challenge in vivo. lincRNA-EPS localizes at regulatory regions of IRGs to control nucleosome positioning and repress transcription. Further, lincRNA-EPS mediates these effects by interacting with heterogeneous nuclear ribonucleoprotein L via a CANACA motif located in its 3' end. Together, these findings identify lincRNA-EPS as a repressor of inflammatory responses, highlighting the importance of lincRNAs in the immune system.


Subject(s)
Gene Expression Regulation , Inflammation/genetics , Macrophages/immunology , RNA, Long Noncoding/metabolism , Animals , Chromatids/metabolism , Gene Deletion , Humans , Listeria monocytogenes/physiology , Listeriosis/immunology , Macrophages/metabolism , Macrophages/microbiology , Macrophages/virology , Mice , Mice, Inbred C57BL , RNA, Long Noncoding/genetics , Respirovirus Infections/immunology , Sendai virus/physiology , Toll-Like Receptors/metabolism , Transcriptome
20.
Proc Natl Acad Sci U S A ; 112(42): E5679-88, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26438848

ABSTRACT

Reactive oxygen species (ROS) such as hydrogen peroxide (H2O2) govern cellular homeostasis by inducing signaling. H2O2 modulates the activity of phosphatases and many other signaling molecules through oxidation of critical cysteine residues, which led to the notion that initiation of ROS signaling is broad and nonspecific, and thus fundamentally distinct from other signaling pathways. Here, we report that H2O2 signaling bears hallmarks of a regular signal transduction cascade. It is controlled by hierarchical signaling events resulting in a focused response as the results place the mitochondrial respiratory chain upstream of tyrosine-protein kinase Lyn, Lyn upstream of tyrosine-protein kinase SYK (Syk), and Syk upstream of numerous targets involved in signaling, transcription, translation, metabolism, and cell cycle regulation. The active mediators of H2O2 signaling colocalize as H2O2 induces mitochondria-associated Lyn and Syk phosphorylation, and a pool of Lyn and Syk reside in the mitochondrial intermembrane space. Finally, the same intermediaries control the signaling response in tissues and species responsive to H2O2 as the respiratory chain, Lyn, and Syk were similarly required for H2O2 signaling in mouse B cells, fibroblasts, and chicken DT40 B cells. Consistent with a broad role, the Syk pathway is coexpressed across tissues, is of early metazoan origin, and displays evidence of evolutionary constraint in the human. These results suggest that H2O2 signaling is under control of a signal transduction pathway that links the respiratory chain to the mitochondrial intermembrane space-localized, ubiquitous, and ancient Syk pathway in hematopoietic and nonhematopoietic cells.


Subject(s)
Electron Transport , Hydrogen Peroxide/metabolism , Mitochondrial Membranes/metabolism , Signal Transduction , Animals , Cells, Cultured , Chickens , Enzyme Activation , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , Syk Kinase , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...