Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
1.
iScience ; 25(1): 103680, 2022 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-35036870

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-34731223

RESUMO

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.


Assuntos
LDL-Colesterol/sangue , Regulação para Baixo , Glicoforinas/genética , Hiperlipidemias/prevenção & controle , Pró-Proteína Convertase 9/sangue , Receptores de LDL/genética , Animais , Peso Corporal , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Eritrócitos/metabolismo , Feminino , Engenharia Genética , Glicoforinas/química , Células HEK293 , Humanos , Hiperlipidemias/induzido quimicamente , Hiperlipidemias/metabolismo , Camundongos , Gravidez , Receptores de LDL/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transplante de Células-Tronco , Transdução Genética
3.
Science ; 371(6532): 867, 2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33632822
4.
Exp Hematol ; 89: 1-12, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32798645

RESUMO

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.


Assuntos
Células Precursoras Eritroides/metabolismo , Hematologia/história , Biologia Molecular/história , Receptores da Eritropoetina/história , Talassemia beta/genética , Clonagem Molecular , Eritrócitos/metabolismo , Eritrócitos/patologia , Células Precursoras Eritroides/citologia , Eritropoese/genética , Expressão Gênica , História do Século XX , História do Século XXI , Humanos , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , alfa-Globinas/genética , alfa-Globinas/metabolismo , Globinas beta/genética , Globinas beta/metabolismo , Talassemia beta/metabolismo , Talassemia beta/patologia
5.
Proc Natl Acad Sci U S A ; 117(26): 15055-15065, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32554489

RESUMO

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.


Assuntos
Tecido Adiposo Marrom/fisiologia , Monoéster Fosfórico Hidrolases/genética , Fosforilcolina/metabolismo , Termogênese , Adipócitos Marrons/enzimologia , Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/enzimologia , Animais , Temperatura Baixa , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monoéster Fosfórico Hidrolases/deficiência
6.
Dev Cell ; 49(1): 118-129.e7, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30827895

RESUMO

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.


Assuntos
Células Sanguíneas/metabolismo , Proliferação de Células/genética , Células Precursoras Eritroides/metabolismo , Hematopoese/genética , Animais , Células Sanguíneas/citologia , Diferenciação Celular/genética , Divisão Celular/genética , Células Cultivadas , Eritrócitos/citologia , Eritrócitos/metabolismo , Células Eritroides/citologia , Células Eritroides/metabolismo , Células Precursoras Eritroides/citologia , Eritropoese/genética , Glucocorticoides/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Camundongos , Análise de Célula Única/métodos , Transcriptoma/genética
7.
J Biol Chem ; 293(51): 19797-19811, 2018 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-30366982

RESUMO

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.


Assuntos
Células Eritroides/metabolismo , Eritropoetina/metabolismo , Ferroquelatase/metabolismo , Heme/biossíntese , Ferro/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Animais , Células Eritroides/citologia , Eritropoese , Células HEK293 , Humanos , Proteínas de Membrana/química , Camundongos , Membranas Mitocondriais/metabolismo , Proteínas Mitocondriais/química , Transporte Proteico
8.
Hematol Oncol Clin North Am ; 32(4): 701-712, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30047421

RESUMO

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.


Assuntos
Anemia de Diamond-Blackfan , Transfusão de Eritrócitos , Transplante de Células-Tronco Hematopoéticas , Mutação , Doadores de Tecidos , Aloenxertos , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Anemia de Diamond-Blackfan/patologia , Anemia de Diamond-Blackfan/terapia , Humanos , Irmãos
9.
Blood ; 131(26): 2955-2966, 2018 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-29712634

RESUMO

Red cells contain a unique constellation of membrane lipids. Although much is known about regulated protein expression, the regulation of lipid metabolism during erythropoiesis is poorly studied. Here, we show that transcription of PHOSPHO1, a phosphoethanolamine and phosphocholine phosphatase that mediates the hydrolysis of phosphocholine to choline, is strongly upregulated during the terminal stages of erythropoiesis of both human and mouse erythropoiesis, concomitant with increased catabolism of phosphatidylcholine (PC) and phosphocholine as shown by global lipidomic analyses of mouse and human terminal erythropoiesis. Depletion of PHOSPHO1 impaired differentiation of fetal mouse and human erythroblasts, and, in adult mice, depletion impaired phenylhydrazine-induced stress erythropoiesis. Loss of PHOSPHO1 also impaired phosphocholine catabolism in mouse fetal liver progenitors and resulted in accumulation of several lipids; adenosine triphosphate (ATP) production was reduced as a result of decreased oxidative phosphorylation. Glycolysis replaced oxidative phosphorylation in PHOSPHO1-knockout erythroblasts and the increased glycolysis was used for the production of serine or glycine. Our study elucidates the dynamic changes in lipid metabolism during terminal erythropoiesis and reveals the key roles of PC and phosphocholine metabolism in energy balance and amino acid supply.


Assuntos
Eritroblastos/metabolismo , Eritropoese , Fosforilcolina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Eritroblastos/citologia , Deleção de Genes , Glicólise , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação Oxidativa , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo
10.
Nat Commun ; 8(1): 2115, 2017 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-29235464

RESUMO

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.


Assuntos
Adipócitos Marrons/enzimologia , Tecido Adiposo Marrom/metabolismo , Diferenciação Celular , Quinase Syk/metabolismo , Adipócitos Marrons/citologia , Animais , Proliferação de Células , Células Cultivadas , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Quinase Syk/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
11.
Mol Biol Cell ; 28(22): 2908-2910, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29084906

RESUMO

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.


Assuntos
Tutoria/tendências , Seleção de Pessoal/métodos , Humanos , Laboratórios , Mentores , Pesquisadores
12.
Blood ; 130(18): 1965-1975, 2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-28928124

RESUMO

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.


Assuntos
Neoplasias Hematológicas/genética , Hematopoese/genética , RNA Longo não Codificante/genética , Animais , Desenvolvimento Embrionário/genética , Humanos , Modelos Biológicos , Proteínas/metabolismo , RNA Longo não Codificante/metabolismo
13.
Nat Commun ; 8(1): 647, 2017 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-28935898

RESUMO

Programmed death ligand 1 (PD-L1) is expressed on a number of immune and cancer cells, where it can downregulate antitumor immune responses. Its expression has been linked to metabolic changes in these cells. Here we develop a radiolabeled camelid single-domain antibody (anti-PD-L1 VHH) to track PD-L1 expression by immuno-positron emission tomography (PET). PET-CT imaging shows a robust and specific PD-L1 signal in brown adipose tissue (BAT). We confirm expression of PD-L1 on brown adipocytes and demonstrate that signal intensity does not change in response to cold exposure or ß-adrenergic activation. This is the first robust method of visualizing murine brown fat independent of its activation state.Current approaches to visualise brown adipose tissue (BAT) rely primarily on markers that reflect its metabolic activity. Here, the authors show that PD-L1 is expressed on brown adipocytes, does not change upon BAT activation, and that BAT volume in mice can be measured by PET-CT with a radiolabeled anti-PD-L1 antibody.


Assuntos
Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/metabolismo , Antígeno B7-H1/análise , Biomarcadores/análise , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/diagnóstico por imagem , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/imunologia , Camelídeos Americanos/imunologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada/métodos , Reprodutibilidade dos Testes
14.
Proc Natl Acad Sci U S A ; 114(38): 10107-10112, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28864529

RESUMO

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.


Assuntos
Diferenciação Celular , Coativadores de Receptor Nuclear/metabolismo , Reticulócitos/metabolismo , Receptores beta dos Hormônios Tireóideos/metabolismo , Hormônios Tireóideos/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Estudo de Associação Genômica Ampla , Humanos , Camundongos , Camundongos Knockout , Coativadores de Receptor Nuclear/genética , Receptores beta dos Hormônios Tireóideos/genética , Hormônios Tireóideos/genética
15.
Nat Commun ; 8(1): 423, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28871080

RESUMO

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.


Assuntos
Toxinas Botulínicas Tipo A/toxicidade , Eritrócitos/metabolismo , Engenharia Genética , Anticorpos de Domínio Único/genética , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/metabolismo , Toxinas Botulínicas Tipo A/metabolismo , Botulismo/etiologia , Botulismo/terapia , Transfusão de Eritrócitos , Eritrócitos/virologia , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/transplante , Células Precursoras Eritroides/virologia , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glicoforinas/genética , Glicoforinas/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Retroviridae/genética , Retroviridae/metabolismo , Anticorpos de Domínio Único/administração & dosagem , Anticorpos de Domínio Único/metabolismo
16.
Cell Rep ; 19(12): 2503-2514, 2017 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-28636939

RESUMO

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.


Assuntos
Eritrócitos/fisiologia , Eritropoese , RNA Longo não Codificante/fisiologia , Animais , Células Cultivadas , Elementos Facilitadores Genéticos , Células Eritroides/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Transcrição Gênica
17.
Elife ; 62017 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-28553927

RESUMO

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.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Eritropoetina/metabolismo , Fator de Transcrição GATA1/metabolismo , Heme/biossíntese , Transdução de Sinais , Animais , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Camundongos , Membranas Mitocondriais/metabolismo , Peixe-Zebra
18.
Proc Natl Acad Sci U S A ; 114(12): 3157-3162, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28270614

RESUMO

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.

19.
Nat Rev Mol Cell Biol ; 18(4): 214, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28248321
20.
Sci Transl Med ; 9(376)2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28179501

RESUMO

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.


Assuntos
Anemia de Diamond-Blackfan/tratamento farmacológico , Descoberta de Drogas , Células-Tronco Hematopoéticas/metabolismo , Compostos Alílicos/farmacologia , Anemia de Diamond-Blackfan/patologia , Antígenos CD34/metabolismo , Autofagia/efeitos dos fármacos , Proteína 5 Relacionada à Autofagia/metabolismo , Diferenciação Celular/efeitos dos fármacos , Reprogramação Celular , Células Eritroides/efeitos dos fármacos , Células Eritroides/patologia , Eritropoese/efeitos dos fármacos , Teste de Complementação Genética , Globinas/metabolismo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Quinazolinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...