Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 161(4): 858-67, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25957689

RESUMEN

The mitochondrion maintains and regulates its proteome with chaperones primarily inherited from its bacterial endosymbiont ancestor. Among these chaperones is the AAA+ unfoldase ClpX, an important regulator of prokaryotic physiology with poorly defined function in the eukaryotic mitochondrion. We observed phenotypic similarity in S. cerevisiae genetic interaction data between mitochondrial ClpX (mtClpX) and genes contributing to heme biosynthesis, an essential mitochondrial function. Metabolomic analysis revealed that 5-aminolevulinic acid (ALA), the first heme precursor, is 5-fold reduced in yeast lacking mtClpX activity and that total heme is reduced by half. mtClpX directly stimulates ALA synthase in vitro by catalyzing incorporation of its cofactor, pyridoxal phosphate. This activity is conserved in mammalian homologs; additionally, mtClpX depletion impairs vertebrate erythropoiesis, which requires massive upregulation of heme biosynthesis to supply hemoglobin. mtClpX, therefore, is a widely conserved stimulator of an essential biosynthetic pathway and uses a previously unrecognized mechanism for AAA+ unfoldases.


Asunto(s)
Endopeptidasa Clp/metabolismo , Eritropoyesis , Eucariontes/metabolismo , Hemo/biosíntesis , 5-Aminolevulinato Sintetasa/metabolismo , Secuencia de Aminoácidos , Ácido Aminolevulínico/metabolismo , Animales , Evolución Biológica , Endopeptidasa Clp/química , Endopeptidasa Clp/genética , Eucariontes/genética , Humanos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares/metabolismo , Datos de Secuencia Molecular , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Alineación de Secuencia , Pez Cebra/metabolismo
2.
J Biol Chem ; 295(32): 11002-11020, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32518166

RESUMEN

Mitochondrial iron import is essential for iron-sulfur cluster formation and heme biosynthesis. Two nuclear-encoded vertebrate mitochondrial high-affinity iron importers, mitoferrin1 (Mfrn1) and Mfrn2, have been identified in mammals. In mice, the gene encoding Mfrn1, solute carrier family 25 member 37 (Slc25a37), is highly expressed in sites of erythropoiesis, and whole-body Slc25a37 deletion leads to lethality. Here, we report that mice with a deletion of Slc25a28 (encoding Mfrn2) are born at expected Mendelian ratios, but show decreased male fertility due to reduced sperm numbers and sperm motility. Mfrn2-/- mice placed on a low-iron diet exhibited reduced mitochondrial manganese, cobalt, and zinc levels, but not reduced iron. Hepatocyte-specific loss of Slc25a37 (encoding Mfrn1) in Mfrn2-/- mice did not affect animal viability, but resulted in a 40% reduction in mitochondrial iron and reduced levels of oxidative phosphorylation proteins. Placing animals on a low-iron diet exaggerated the reduction in mitochondrial iron observed in liver-specific Mfrn1/2-knockout animals. Mfrn1-/-/Mfrn2-/- bone marrow-derived macrophages or skin fibroblasts in vitro were unable to proliferate, and overexpression of Mfrn1-GFP or Mfrn2-GFP prevented this proliferation defect. Loss of both mitoferrins in hepatocytes dramatically reduced regeneration in the adult mouse liver, further supporting the notion that both mitoferrins transport iron and that their absence limits proliferative capacity of mammalian cells. We conclude that Mfrn1 and Mfrn2 contribute to mitochondrial iron homeostasis and are required for high-affinity iron import during active proliferation of mammalian cells.


Asunto(s)
Proteínas de Transporte de Catión/fisiología , Proliferación Celular/fisiología , Regeneración Hepática/fisiología , Proteínas de Transporte de Membrana/fisiología , Animales , Homeostasis , Hierro/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Hepáticas/metabolismo
3.
Proc Natl Acad Sci U S A ; 114(38): E8045-E8052, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28874591

RESUMEN

Loss-of-function mutations in genes for heme biosynthetic enzymes can give rise to congenital porphyrias, eight forms of which have been described. The genetic penetrance of the porphyrias is clinically variable, underscoring the role of additional causative, contributing, and modifier genes. We previously discovered that the mitochondrial AAA+ unfoldase ClpX promotes heme biosynthesis by activation of δ-aminolevulinate synthase (ALAS), which catalyzes the first step of heme synthesis. CLPX has also been reported to mediate heme-induced turnover of ALAS. Here we report a dominant mutation in the ATPase active site of human CLPX, p.Gly298Asp, that results in pathological accumulation of the heme biosynthesis intermediate protoporphyrin IX (PPIX). Amassing of PPIX in erythroid cells promotes erythropoietic protoporphyria (EPP) in the affected family. The mutation in CLPX inactivates its ATPase activity, resulting in coassembly of mutant and WT protomers to form an enzyme with reduced activity. The presence of low-activity CLPX increases the posttranslational stability of ALAS, causing increased ALAS protein and ALA levels, leading to abnormal accumulation of PPIX. Our results thus identify an additional molecular mechanism underlying the development of EPP and further our understanding of the multiple mechanisms by which CLPX controls heme metabolism.


Asunto(s)
5-Aminolevulinato Sintetasa/metabolismo , Endopeptidasa Clp , Mutación Missense , Porfiria Eritropoyética , Protoporfirinas/biosíntesis , 5-Aminolevulinato Sintetasa/genética , Adolescente , Sustitución de Aminoácidos , Endopeptidasa Clp/genética , Endopeptidasa Clp/metabolismo , Estabilidad de Enzimas/genética , Femenino , Humanos , Masculino , Porfiria Eritropoyética/genética , Porfiria Eritropoyética/metabolismo , Porfiria Eritropoyética/patología , Protoporfirinas/genética
4.
J Biol Chem ; 293(51): 19797-19811, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30366982

RESUMEN

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.


Asunto(s)
Células Eritroides/metabolismo , Eritropoyetina/metabolismo , Ferroquelatasa/metabolismo , Hemo/biosíntesis , Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Células Eritroides/citología , Eritropoyesis , Células HEK293 , Humanos , Proteínas de la Membrana/química , Ratones , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/química , Transporte de Proteínas
5.
J Biol Chem ; 292(39): 16284-16299, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28808058

RESUMEN

ATP-binding cassette subfamily B member 10 (Abcb10) is a mitochondrial ATP-binding cassette (ABC) transporter that complexes with mitoferrin1 and ferrochelatase to enhance heme biosynthesis in developing red blood cells. Reductions in Abcb10 levels have been shown to reduce mitoferrin1 protein levels and iron import into mitochondria, resulting in reduced heme biosynthesis. As an ABC transporter, Abcb10 binds and hydrolyzes ATP, but its transported substrate is unknown. Here, we determined that decreases in Abcb10 did not result in protoporphyrin IX accumulation in morphant-treated zebrafish embryos or in differentiated Abcb10-specific shRNA murine Friend erythroleukemia (MEL) cells in which Abcb10 was specifically silenced with shRNA. We also found that the ATPase activity of Abcb10 is necessary for hemoglobinization in MEL cells, suggesting that the substrate transported by Abcb10 is important in mediating increased heme biosynthesis during erythroid development. Inhibition of 5-aminolevulinic acid dehydratase (EC 4.2.1.24) with succinylacetone resulted in both 5-aminolevulinic acid (ALA) accumulation in control and Abcb10-specific shRNA MEL cells, demonstrating that reductions in Abcb10 do not affect ALA export from mitochondria and indicating that Abcb10 does not transport ALA. Abcb10 silencing resulted in an alteration in the heme biosynthesis transcriptional profile due to repression by the transcriptional regulator Bach1, which could be partially rescued by overexpression of Alas2 or Gata1, providing a mechanistic explanation for why Abcb10 shRNA MEL cells exhibit reduced hemoglobinization. In conclusion, our findings rule out that Abcb10 transports ALA and indicate that Abcb10's ATP-hydrolysis activity is critical for hemoglobinization and that the substrate transported by Abcb10 provides a signal that optimizes hemoglobinization.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Regulación Enzimológica de la Expresión Génica , Hemo/biosíntesis , Proteínas de Pez Cebra/metabolismo , Transportadoras de Casetes de Unión a ATP/antagonistas & inhibidores , Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/genética , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/antagonistas & inhibidores , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Embrión no Mamífero/enzimología , Embrión no Mamífero/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Microinyecciones , Morfolinos/metabolismo , Mutación , Interferencia de ARN , ARN Interferente Pequeño , Pez Cebra , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética
6.
Blood ; 128(15): 1913-1917, 2016 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-27488349

RESUMEN

The congenital sideroblastic anemias (CSAs) are a heterogeneous group of inherited blood disorders characterized by pathological mitochondrial iron deposition in erythroid precursors. Each known cause has been attributed to a mutation in a protein associated with heme biosynthesis, iron-sulfur cluster biogenesis, mitochondrial translation, or a component of the mitochondrial respiratory chain. Here, we describe a recurring mutation, c.276_278del, p.F93del, in NDUFB11, a mitochondrial respiratory complex I-associated protein encoded on the X chromosome, in 5 males with a variably syndromic, normocytic CSA. The p.F93del mutation results in respiratory insufficiency and loss of complex I stability and activity in patient-derived fibroblasts. Targeted introduction of this allele into K562 erythroleukemia cells results in a proliferation defect with minimal effect on erythroid differentiation potential, suggesting the mechanism of anemia in this disorder.


Asunto(s)
Anemia Sideroblástica/genética , Secuencia de Bases , Cromosomas Humanos X/genética , Complejo I de Transporte de Electrón/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Eliminación de Secuencia , Adolescente , Adulto , Anciano , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patología , Niño , Preescolar , Cromosomas Humanos X/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Humanos , Células K562 , Masculino , Persona de Mediana Edad
7.
Nature ; 491(7425): 608-12, 2012 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-23135403

RESUMEN

Defects in the availability of haem substrates or the catalytic activity of the terminal enzyme in haem biosynthesis, ferrochelatase (Fech), impair haem synthesis and thus cause human congenital anaemias. The interdependent functions of regulators of mitochondrial homeostasis and enzymes responsible for haem synthesis are largely unknown. To investigate this we used zebrafish genetic screens and cloned mitochondrial ATPase inhibitory factor 1 (atpif1) from a zebrafish mutant with profound anaemia, pinotage (pnt (tq209)). Here we describe a direct mechanism establishing that Atpif1 regulates the catalytic efficiency of vertebrate Fech to synthesize haem. The loss of Atpif1 impairs haemoglobin synthesis in zebrafish, mouse and human haematopoietic models as a consequence of diminished Fech activity and elevated mitochondrial pH. To understand the relationship between mitochondrial pH, redox potential, [2Fe-2S] clusters and Fech activity, we used genetic complementation studies of Fech constructs with or without [2Fe-2S] clusters in pnt, as well as pharmacological agents modulating mitochondrial pH and redox potential. The presence of [2Fe-2S] cluster renders vertebrate Fech vulnerable to perturbations in Atpif1-regulated mitochondrial pH and redox potential. Therefore, Atpif1 deficiency reduces the efficiency of vertebrate Fech to synthesize haem, resulting in anaemia. The identification of mitochondrial Atpif1 as a regulator of haem synthesis advances our understanding of the mechanisms regulating mitochondrial haem homeostasis and red blood cell development. An ATPIF1 deficiency may contribute to important human diseases, such as congenital sideroblastic anaemias and mitochondriopathies.


Asunto(s)
Eritroblastos/metabolismo , Eritropoyesis , Hemo/biosíntesis , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Proteínas/metabolismo , Anemia Sideroblástica/genética , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patología , Animales , Modelos Animales de Enfermedad , Eritroblastos/citología , Ferroquelatasa/metabolismo , Prueba de Complementación Genética , Humanos , Concentración de Iones de Hidrógeno , Ratones , Mitocondrias/patología , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/genética , Oxidación-Reducción , Proteínas/genética , Pez Cebra/metabolismo , Proteína Inhibidora ATPasa
8.
Genes Dev ; 24(15): 1620-33, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20679398

RESUMEN

The bicistronic microRNA (miRNA) locus miR-144/451 is highly expressed during erythrocyte development, although its physiological roles are poorly understood. We show that miR-144/451 ablation in mice causes mild erythrocyte instability and increased susceptibility to damage after exposure to oxidant drugs. This phenotype is deeply conserved, as miR-451 depletion synergizes with oxidant stress to cause profound anemia in zebrafish embryos. At least some protective activities of miR-451 stem from its ability to directly suppress production of 14-3-3zeta, a phospho-serine/threonine-binding protein that inhibits nuclear accumulation of transcription factor FoxO3, a positive regulator of erythroid anti-oxidant genes. Thus, in miR-144/451(-/-) erythroblasts, 14-3-3zeta accumulates, causing partial relocalization of FoxO3 from nucleus to cytoplasm with dampening of its transcriptional program, including anti-oxidant-encoding genes Cat and Gpx1. Supporting this mechanism, overexpression of 14-3-3zeta in erythroid cells and fibroblasts inhibits nuclear localization and activity of FoxO3. Moreover, shRNA suppression of 14-3-3zeta protects miR-144/451(-/-) erythrocytes against peroxide-induced destruction, and restores catalase activity. Our findings define a novel miRNA-regulated pathway that protects erythrocytes against oxidant stress, and, more generally, illustrate how a miRNA can influence gene expression by altering the activity of a key transcription factor.


Asunto(s)
Proteínas 14-3-3/metabolismo , Células Eritroides/metabolismo , Regulación del Desarrollo de la Expresión Génica , MicroARNs/metabolismo , Estrés Oxidativo , Proteínas 14-3-3/genética , Transporte Activo de Núcleo Celular , Animales , Secuencia de Bases , Catalasa/metabolismo , Células Eritroides/enzimología , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/metabolismo , Técnicas de Silenciamiento del Gen , Ratones , Ratones Noqueados , MicroARNs/genética , Alineación de Secuencia , Eliminación de Secuencia/genética , Pez Cebra/genética , Pez Cebra/metabolismo
9.
Blood ; 126(26): 2811-20, 2015 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-26385351

RESUMEN

Rare endothelial cells in the aorta-gonad-mesonephros (AGM) transition into hematopoietic stem cells (HSCs) during embryonic development. Lineage tracing experiments indicate that HSCs emerge from cadherin 5 (Cdh5; vascular endothelial-cadherin)(+) endothelial precursors, and isolated populations of Cdh5(+) cells from mouse embryos and embryonic stem cells can be differentiated into hematopoietic cells. Cdh5 has also been widely implicated as a marker of AGM-derived hemogenic endothelial cells. Because Cdh5(-/-) mice embryos die before the first HSCs emerge, it is unknown whether Cdh5 has a direct role in HSC emergence. Our previous genetic screen yielded malbec (mlb(bw306)), a zebrafish mutant for cdh5, with normal embryonic and definitive blood. Using time-lapse confocal imaging, parabiotic surgical pairing of zebrafish embryos, and blastula transplantation assays, we show that HSCs emerge, migrate, engraft, and differentiate in the absence of cdh5 expression. By tracing Cdh5(-/-)green fluorescent protein (GFP)(+/+) cells in chimeric mice, we demonstrated that Cdh5(-/-)GFP(+/+) HSCs emerging from embryonic day 10.5 and 11.5 (E10.5 and E11.5) AGM or derived from E13.5 fetal liver not only differentiate into hematopoietic colonies but also engraft and reconstitute multilineage adult blood. We also developed a conditional mouse Cdh5 knockout (Cdh5(flox/flox):Scl-Cre-ER(T)) and demonstrated that multipotent hematopoietic colonies form despite the absence of Cdh5. These data establish that Cdh5, a marker of hemogenic endothelium in the AGM, is dispensable for the transition of hemogenic endothelium to HSCs.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Diferenciación Celular/fisiología , Hemangioblastos/citología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Animales , Linaje de la Célula/fisiología , Electroporación , Embrión de Mamíferos , Embrión no Mamífero , Citometría de Flujo , Inmunohistoquímica , Mesonefro/embriología , Ratones , Ratones Noqueados , Microscopía Confocal , Pez Cebra
10.
Am J Hum Genet ; 93(5): 906-14, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24119684

RESUMEN

We used exome sequencing to identify mutations in sideroflexin 4 (SFXN4) in two children with mitochondrial disease (the more severe case also presented with macrocytic anemia). SFXN4 is an uncharacterized mitochondrial protein that localizes to the mitochondrial inner membrane. sfxn4 knockdown in zebrafish recapitulated the mitochondrial respiratory defect observed in both individuals and the macrocytic anemia with megaloblastic features of the more severe case. In vitro and in vivo complementation studies with fibroblasts from the affected individuals and zebrafish demonstrated the requirement of SFXN4 for mitochondrial respiratory homeostasis and erythropoiesis. Our findings establish mutations in SFXN4 as a cause of mitochondriopathy and macrocytic anemia.


Asunto(s)
Anemia Macrocítica/genética , Proteínas de la Membrana/genética , Enfermedades Mitocondriales/genética , Adolescente , Animales , Niño , Eritropoyesis/genética , Exoma , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Proteínas Mitocondriales/genética , Mutación , Pez Cebra/genética
11.
PLoS Genet ; 9(6): e1003533, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23754957

RESUMEN

Heterozygous germline mutations and deletions in PHOX2B, a key regulator of autonomic neuron development, predispose to neuroblastoma, a tumor of the peripheral sympathetic nervous system. To gain insight into the oncogenic mechanisms engaged by these changes, we used zebrafish models to study the functional consequences of aberrant PHOX2B expression in the cells of the developing sympathetic nervous system. Allelic deficiency, modeled by phox2b morpholino knockdown, led to a decrease in the terminal differentiation markers th and dbh in sympathetic ganglion cells. The same effect was seen on overexpression of two distinct neuroblastoma-associated frameshift mutations, 676delG and K155X - but not the R100L missense mutation - in the presence of endogenous Phox2b, pointing to their dominant-negative effects. We demonstrate that Phox2b is capable of regulating itself as well as ascl1, and that phox2b deficiency uncouples this autoregulatory mechanism, leading to inhibition of sympathetic neuron differentiation. This effect on terminal differentiation is associated with an increased number of phox2b(+), ascl1(+), elavl3(-) cells that respond poorly to retinoic acid. These findings suggest that a reduced dosage of PHOX2B during development, through either a heterozygous deletion or dominant-negative mutation, imposes a block in the differentiation of sympathetic neuronal precursors, resulting in a cell population that is likely to be susceptible to secondary transforming events.


Asunto(s)
Diferenciación Celular/genética , Proteínas de Homeodominio/genética , Neuroblastoma/genética , Neurogénesis , Factores de Transcripción/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas ELAV/genética , Proteínas ELAV/metabolismo , Proteína 3 Similar a ELAV , Regulación del Desarrollo de la Expresión Génica , Predisposición Genética a la Enfermedad , Heterocigoto , Proteínas de Homeodominio/metabolismo , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patología , Neuronas/citología , Neuronas/metabolismo , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/patología , Factores de Transcripción/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
12.
J Biol Chem ; 289(31): 21312-24, 2014 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-24907273

RESUMEN

The clustered regularly interspaced short [corrected] palindromic repeats (CRISPR)/CRISPR-associated (Cas) 9 nuclease system has provided a powerful tool for genome engineering. Double strand breaks may trigger nonhomologous end joining repair, leading to frameshift mutations, or homology-directed repair using an extrachromosomal template. Alternatively, genomic deletions may be produced by a pair of double strand breaks. The efficiency of CRISPR/Cas9-mediated genomic deletions has not been systematically explored. Here, we present a methodology for the production of deletions in mammalian cells, ranging from 1.3 kb to greater than 1 Mb. We observed a high frequency of intended genomic deletions. Nondeleted alleles are nonetheless often edited with inversions or small insertion/deletions produced at CRISPR recognition sites. Deleted alleles also typically include small insertion/deletions at predicted deletion junctions. We retrieved cells with biallelic deletion at a frequency exceeding that of probabilistic expectation. We demonstrate an inverse relationship between deletion frequency and deletion size. This work suggests that CRISPR/Cas9 is a robust system to produce a spectrum of genomic deletions to allow investigation of genes and genetic elements.


Asunto(s)
Sistemas CRISPR-Cas/fisiología , Eliminación de Gen , Secuencias Repetitivas Esparcidas , Animales , Secuencia de Bases , Línea Celular Tumoral , Genómica , Ratones , Datos de Secuencia Molecular
13.
J Biol Chem ; 289(11): 7835-43, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24509859

RESUMEN

Mitochondrial iron is essential for the biosynthesis of heme and iron-sulfur ([Fe-S]) clusters in mammalian cells. In developing erythrocytes, iron is imported into the mitochondria by MFRN1 (mitoferrin-1, SLC25A37). Although loss of MFRN1 in zebrafish and mice leads to profound anemia, mutant animals showed no overt signs of porphyria, suggesting that mitochondrial iron deficiency does not result in an accumulation of protoporphyrins. Here, we developed a gene trap model to provide in vitro and in vivo evidence that iron regulatory protein-1 (IRP1) inhibits protoporphyrin accumulation. Mfrn1(+/gt);Irp1(-/-) erythroid cells exhibit a significant increase in protoporphyrin levels. IRP1 attenuates protoporphyrin biosynthesis by binding to the 5'-iron response element (IRE) of alas2 mRNA, inhibiting its translation. Ectopic expression of alas2 harboring a mutant IRE, preventing IRP1 binding, in Mfrn1(gt/gt) cells mimics Irp1 deficiency. Together, our data support a model whereby impaired mitochondrial [Fe-S] cluster biogenesis in Mfrn1(gt/gt) cells results in elevated IRP1 RNA-binding that attenuates ALAS2 mRNA translation and protoporphyrin accumulation.


Asunto(s)
5-Aminolevulinato Sintetasa/metabolismo , Regulación de la Expresión Génica , Proteína 1 Reguladora de Hierro/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Porfirias/metabolismo , Animales , Blastocisto/citología , Diferenciación Celular , Línea Celular Tumoral , Femenino , Genotipo , Células HEK293 , Hemo/química , Humanos , Hierro/química , Proteínas Hierro-Azufre/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Biosíntesis de Proteínas , Protoporfirinas/metabolismo , Pez Cebra
14.
Am J Hum Genet ; 90(1): 40-8, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22226084

RESUMEN

Ocular coloboma is a developmental defect of the eye and is due to abnormal or incomplete closure of the optic fissure. This disorder displays genetic and clinical heterogeneity. Using a positional cloning approach, we identified a mutation in the ATP-binding cassette (ABC) transporter ABCB6 in a Chinese family affected by autosomal-dominant coloboma. The Leu811Val mutation was identified in seven affected members of the family and was absent in six unaffected members from three generations. A LOD score of 3.2 at θ = 0 was calculated for the mutation identified in this family. Sequence analysis was performed on the ABCB6 exons from 116 sporadic cases of microphthalmia with coloboma (MAC), isolated coloboma, and aniridia, and an additional mutation (A57T) was identified in three patients with MAC. These two mutations were not present in the ethnically matched control populations. Immunostaining of transiently transfected, Myc-tagged ABCB6 in retinal pigment epithelial (RPE) cells showed that it localized to the endoplasmic reticulum and Golgi apparatus of RPE cells. RT-PCR of ABCB6 mRNA in human cell lines and tissue indicated that ABCB6 is expressed in the retinae and RPE cells. Using zebrafish, we show that abcb6 is expressed in the eye and CNS. Morpholino knockdown of abcb6 in zebrafish produces a phenotype characteristic of coloboma and replicates the clinical phenotype observed in our index cases. The knockdown phenotype can be corrected with coinjection of the wild-type, but not mutant, ABCB6 mRNA, suggesting that the phenotypes observed in zebrafish are due to insufficient abcb6 function. Our results demonstrate that ABCB6 mutations cause ocular coloboma.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Coloboma/genética , Mutación , Transportadoras de Casetes de Unión a ATP/biosíntesis , Animales , Pueblo Asiatico/genética , Secuencia de Bases , Línea Celular , Sistema Nervioso Central/metabolismo , Exones , Anomalías del Ojo/genética , Femenino , Humanos , Escala de Lod , Masculino , Microftalmía/genética , Persona de Mediana Edad , Datos de Secuencia Molecular , Morfolinos/administración & dosificación , Epitelio Pigmentado de la Retina , Transfección , Pez Cebra , Proteínas de Pez Cebra/genética
15.
Genome Res ; 22(8): 1541-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22555591

RESUMEN

Genetic mapping of mutations in model systems has facilitated the identification of genes contributing to fundamental biological processes including human diseases. However, this approach has historically required the prior characterization of informative markers. Here we report a fast and cost-effective method for genetic mapping using next-generation sequencing that combines single nucleotide polymorphism discovery, mutation localization, and potential identification of causal sequence variants. In contrast to prior approaches, we have developed a hidden Markov model to narrowly define the mutation area by inferring recombination breakpoints of chromosomes in the mutant pool. In addition, we created an interactive online software resource to facilitate automated analysis of sequencing data and demonstrate its utility in the zebrafish and mouse models. Our novel methodology and online tools will make next-generation sequencing an easily applicable resource for mutation mapping in all model systems.


Asunto(s)
Análisis Mutacional de ADN/métodos , Programas Informáticos , Pez Cebra/genética , Alelos , Animales , Mapeo Cromosómico/métodos , Cromosomas/genética , Cruzamientos Genéticos , Femenino , Frecuencia de los Genes , Genómica/métodos , Homocigoto , Masculino , Cadenas de Markov , Ratones , Ratones Endogámicos C57BL , Mutación , Polimorfismo de Nucleótido Simple , Recombinación Genética , Factores de Tiempo
16.
Blood ; 121(13): 2483-93, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23341543

RESUMEN

Many pathways regulating blood formation have been elucidated, yet how each coordinates with embryonic biophysiology to modulate the spatiotemporal production of hematopoietic stem cells (HSCs) is currently unresolved. Here, we report that glucose metabolism impacts the onset and magnitude of HSC induction in vivo. In zebrafish, transient elevations in physiological glucose levels elicited dose-dependent effects on HSC development, including enhanced runx1 expression and hematopoietic cluster formation in the aorta-gonad-mesonephros region; embryonic-to-adult transplantation studies confirmed glucose increased functional HSCs. Glucose uptake was required to mediate the enhancement in HSC development; likewise, metabolic inhibitors diminished nascent HSC production and reversed glucose-mediated effects on HSCs. Increased glucose metabolism preferentially impacted hematopoietic and vascular targets, as determined by gene expression analysis, through mitochondrial-derived reactive oxygen species (ROS)-mediated stimulation of hypoxia-inducible factor 1α (hif1α). Epistasis assays demonstrated that hif1α regulates HSC formation in vivo and mediates the dose-dependent effects of glucose metabolism on the timing and magnitude of HSC production. We propose that this fundamental metabolic-sensing mechanism enables the embryo to respond to changes in environmental energy input and adjust hematopoietic output to maintain embryonic growth and ensure viability.


Asunto(s)
Metabolismo de los Hidratos de Carbono/fisiología , Inducción Embrionaria , Glucosa/metabolismo , Células Madre Hematopoyéticas/fisiología , Animales , Animales Modificados Genéticamente , Metabolismo de los Hidratos de Carbono/genética , Proliferación Celular/efectos de los fármacos , Embrión no Mamífero , Inducción Embrionaria/efectos de los fármacos , Inducción Embrionaria/genética , Regulación del Desarrollo de la Expresión Génica , Glucosa/farmacología , Glucólisis/efectos de los fármacos , Glucólisis/genética , Glucólisis/fisiología , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Fosforilación Oxidativa , Factores de Tiempo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo
17.
Circ Res ; 113(3): 279-87, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23720443

RESUMEN

RATIONALE: Heme plays a critical role in gas exchange, mitochondrial energy production, and antioxidant defense in cardiovascular system. The mitochondrial transporter ATP-binding cassette (ABC) B10 has been suggested to export heme out of the mitochondria and is required for normal hemoglobinization of erythropoietic cells and protection against ischemia-reperfusion injury in the heart; however, its primary function has not been established. OBJECTIVE: The aim of this study was to identify the function of ABCB10 in heme synthesis in cardiac cells. METHODS AND RESULTS: Knockdown of ABCB10 in cardiac myoblasts significantly reduced heme levels and the activities of heme-containing proteins, whereas supplementation with δ-aminolevulinic acid reversed these defects. Overexpression of mitochondrial δ-aminolevulinic acid synthase 2, the rate-limiting enzyme upstream of δ-aminolevulinic acid export, failed to restore heme levels in cells with ABCB10 downregulation. ABCB10 and heme levels were increased by hypoxia, and reversal of ABCB10 upregulation caused oxidative stress and cell death. Furthermore, ABCB10 knockdown in neonatal rat cardiomyocytes resulted in a significant delay of calcium removal from the cytoplasm, suggesting a relaxation defect. Finally, ABCB10 expression and heme levels were altered in failing human hearts and mice with ischemic cardiomyopathy. CONCLUSIONS: ABCB10 plays a critical role in heme synthesis pathway by facilitating δ-aminolevulinic acid production or export from the mitochondria. In contrast to previous reports, we show that ABCB10 is not a heme exporter and instead is required for the early mitochondrial steps of heme biosynthesis.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Hemo/biosíntesis , Mitocondrias Cardíacas/fisiología , Miocitos Cardíacos/fisiología , Animales , Células Cultivadas , Hemo/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratas
18.
Proc Natl Acad Sci U S A ; 109(30): 12099-104, 2012 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-22773809

RESUMEN

Phenotype-driven approaches to gene discovery using inbred mice have been instrumental in identifying genetic determinants of inherited blood dyscrasias. The recessive mutant scat (severe combined anemia and thrombocytopenia) alternates between crisis and remission episodes, indicating an aberrant regulatory feedback mechanism common to erythrocyte and platelet formation. Here, we identify a missense mutation (G125V) in the scat Rasa3 gene, encoding a Ras GTPase activating protein (RasGAP), and elucidate the mechanism producing crisis episodes. The mutation causes mislocalization of RASA3 to the cytosol in scat red cells where it is inactive, leading to increased GTP-bound Ras. Erythropoiesis is severely blocked in scat crisis mice, and ~94% succumb during the second crisis (~30 d of age) from catastrophic hematopoietic failure in the spleen and bone marrow. Megakaryopoiesis is also defective during crisis. Notably, the scat phenotype is recapitulated in zebrafish when rasa3 is silenced. These results highlight a critical, conserved, and nonredundant role for RASA3 in vertebrate hematopoiesis.


Asunto(s)
Eritropoyesis/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Trombopoyesis/fisiología , Animales , Animales Modificados Genéticamente , Activación Enzimática/fisiología , Eritropoyesis/genética , GTP Fosfohidrolasas/metabolismo , Ratones , Mutación Missense/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Trombopoyesis/genética , Pez Cebra
19.
Dev Biol ; 373(2): 431-41, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22960038

RESUMEN

Growth Factor Independence (Gfi) transcription factors play essential roles in hematopoiesis, differentially activating and repressing transcriptional programs required for hematopoietic stem/progenitor cell (HSPC) development and lineage specification. In mammals, Gfi1a regulates hematopoietic stem cells (HSC), myeloid and lymphoid populations, while its paralog, Gfi1b, regulates HSC, megakaryocyte and erythroid development. In zebrafish, gfi1aa is essential for primitive hematopoiesis; however, little is known about the role of gfi1aa in definitive hematopoiesis or about additional gfi factors in zebrafish. Here, we report the isolation and characterization of an additional hematopoietic gfi factor, gfi1b. We show that gfi1aa and gfi1b are expressed in the primitive and definitive sites of hematopoiesis in zebrafish. Our functional analyses demonstrate that gfi1aa and gfi1b have distinct roles in regulating primitive and definitive hematopoietic progenitors, respectively. Loss of gfi1aa silences markers of early primitive progenitors, scl and gata1. Conversely, loss of gfi1b silences runx-1, c-myb, ikaros and cd41, indicating that gfi1b is required for definitive hematopoiesis. We determine the epistatic relationships between the gfi factors and key hematopoietic transcription factors, demonstrating that gfi1aa and gfi1b join lmo2, scl, runx-1 and c-myb as critical regulators of teleost HSPC. Our studies establish a comparative paradigm for the regulation of hematopoietic lineages by gfi transcription factors.


Asunto(s)
Proteínas de Unión al ADN/genética , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Clonación Molecular , Secuencia Conservada/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Embrión no Mamífero/metabolismo , Epistasis Genética , Eritropoyesis/genética , Evolución Molecular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Sistema Hematopoyético/embriología , Sistema Hematopoyético/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Pez Cebra/embriología , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/metabolismo
20.
Pflugers Arch ; 466(8): 1605-18, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24668450

RESUMEN

The zebrafish genome encodes two slc4a1 genes, one expressed in erythroid tissues and the other in the HR (H(+)-ATPase-rich) type of embryonic skin ionocytes, and two slc4a2 genes, one in proximal pronephric duct and the other in several extrarenal tissues of the embryo. We now report cDNA cloning and functional characterization of zebrafish slc4a3/ae3 gene products. The single ae3 gene on chromosome 9 generates at least two low-abundance ae3 transcripts differing only in their 5'-untranslated regions and encoding a single definitive Ae3 polypeptide of 1170 amino acids. The 7 kb upstream of the apparent initiator Met in ae3 exon 3 comprises multiple diverse, mobile repeat elements which disrupt and appear to truncate the Ae3 N-terminal amino acid sequence that would otherwise align with brain Ae3 of other species. Embryonic ae3 mRNA expression was detected by whole mount in situ hybridization only in fin buds at 24-72 hpf, but was detectable by RT-PCR across a range of embryonic and adult tissues. Epitope-tagged Ae3 polypeptide was expressed at or near the surface of Xenopus oocytes, and mediated low rates of DIDS-sensitive (36)Cl(-)/Cl(-) exchange in influx and efflux assays. As previously reported for Ae2 polypeptides, (36)Cl(-) transport by Ae3 was inhibited by both extracellular and intracellular acidic pH, and stimulated by alkaline pH. However, zebrafish Ae3 differed from Ae2 polypeptides in its insensitivity to NH4Cl and to hypertonicity. We conclude that multiple repeat elements have disrupted the 5'-end of the zebrafish ae3 gene, associated with N-terminal truncation of the protein and reduced anion transport activity.


Asunto(s)
Antiportadores de Cloruro-Bicarbonato/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Antiportadores de Cloruro-Bicarbonato/metabolismo , Clonación Molecular , Humanos , Hibridación in Situ , Microscopía Confocal , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA