Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2839: 225-231, 2024.
Article in English | MEDLINE | ID: mdl-39008256

ABSTRACT

Radiolabeling enables the quantitation of newly synthesized heme and porphyrin, allowing us to distinguish heme synthesis rates from total cellular heme. Here, we describe a protocol for labeling heme with 14C-glycine or ALA and the sequential extraction of heme and porphyrin from the same samples for quantitation by liquid scintillation.


Subject(s)
Aminolevulinic Acid , Carbon Radioisotopes , Glycine , Heme , Porphyrins , Heme/chemistry , Aminolevulinic Acid/chemistry , Aminolevulinic Acid/metabolism , Carbon Radioisotopes/chemistry , Porphyrins/chemistry , Glycine/chemistry , Isotope Labeling/methods , Humans
2.
bioRxiv ; 2023 Sep 27.
Article in English | MEDLINE | ID: mdl-37823037

ABSTRACT

Iron plays a central role in cellular redox processes, but its ability to adopt multiple oxidation states also enables it to catalyze deleterious reactions. The requirement for iron in erythropoiesis has necessitated the evolution of mechanisms with which to handle the iron required for hemoglobinization. FAM210B was identified as a regulator of mitochondrial iron import and heme synthesis in erythroid cell culture and zebrafish models. In this manuscript, we demonstrate that while FAM210B is required for erythroid differentiation and heme synthesis under standard cell culture conditions, holotransferrin supplementation was sufficient to chemically complement the iron-deficient phenotype. As the biology of FAM210B is complex and context specific, and whole-organism studies on FAM210 proteins have been limited, we sought to unravel the role of FAM210B in erythropoiesis using knockout mice. We were surprised to discover that Fam210b -/- mice were viable and the adults did not have erythropoietic defects in the bone marrow. In contrast to studies in C. elegans, Fam210b -/- mice were also fertile. There were some modest phenotypes, such as a slight increase in lymphocytes and white cell count in Fam210b -/- females, as well as an increase in body weight in Fam210b -/- males. However, our findings suggest that FAM210B may play a more important role in cellular iron homeostasis under iron deficient conditions. Here, we will discuss the cell culture conditions used in iron metabolism studies that can account for the disparate finding on FAM210B function. Moving forward, resolving these discrepancies will be important in identifying novel iron homeostasis genes.

3.
J Biol Chem ; 298(10): 102404, 2022 10.
Article in English | MEDLINE | ID: mdl-35988646

ABSTRACT

Endothelial cells (ECs) are the primary cellular constituent of blood vessels that are in direct contact with hemodynamic forces over their lifetime. Throughout the body, vessels experience different blood flow patterns and rates that alter vascular architecture and cellular behavior. Because of the complexities of studying blood flow in an intact organism, particularly during development, the field has increasingly relied on in vitro modeling of blood flow as a powerful technique for studying hemodynamic-dependent signaling mechanisms in ECs. While commercial flow systems that recirculate fluids exist, many commercially available pumps are peristaltic and best model pulsatile flow conditions. However, there are many important situations in which ECs experience laminar flow conditions in vivo, such as along long straight stretches of the vasculature. To understand EC function under these contexts, it is important to be able to reproducibly model laminar flow conditions in vitro. Here, we outline a method to reliably adapt commercially available peristaltic pumps to study laminar flow conditions. Our proof-of-concept study focuses on 2D models but could be further adapted to 3D environments to better model in vivo scenarios, such as organ development. Our studies make significant inroads into solving technical challenges associated with flow modeling and allow us to conduct functional studies toward understanding the mechanistic role of shear forces on vascular architecture, cellular behavior, and remodeling in diverse physiological contexts.


Subject(s)
Adaptation, Physiological , Endothelial Cells , Endothelial Cells/physiology , Stress, Mechanical , Cells, Cultured
4.
J Biol Chem ; 298(8): 102225, 2022 08.
Article in English | MEDLINE | ID: mdl-35780836

ABSTRACT

Ephrin-B signaling has been implicated in many normal and pathological processes, including neural crest development and tumor metastasis. We showed previously that proteolysis of ephrin-B ligands by the disintegrin metalloprotease ADAM13 is necessary for canonical Wnt signal activation and neural crest induction in Xenopus, but it was unclear if these mechanisms are conserved in mammals. Here, we report that mammalian ADAM9 cleaves ephrin-B1 and ephrin-B2 and can substitute for Xenopus ADAM13 to induce the neural crest. We found that ADAM9 expression is elevated in human colorectal cancer (CRC) tissues and that knockdown (KD) of ADAM9 inhibits the migration and invasion of SW620 and HCT116 CRC cells by reducing the activity of Akt kinase, which is antagonized by ephrin-Bs. Akt is a signaling node that activates multiple downstream pathways, including the Wnt and mTOR pathways, both of which can promote CRC cell migration/invasion. Surprisingly, we also found that KD of ADAM9 downregulates Wnt signaling but has negligible effects on mTOR signaling in SW620 cells; in contrast, mTOR activity is suppressed while Wnt signaling remains unaffected by ADAM9 KD in HCT116 cells. These results suggest that mammalian ADAM9 cleaves ephrin-Bs to derepress Akt and promote CRC migration and invasion; however, the signaling pathways downstream of Akt are differentially regulated by ADAM9 in different CRC cell lines, reflecting the heterogeneity of CRC cells in responding to manipulations of upstream Akt regulators.


Subject(s)
ADAM Proteins/metabolism , Colorectal Neoplasms , Ephrins , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/pathology , Humans , Ligands , Mammals/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Metalloproteases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Wnt Signaling Pathway
5.
Front Cell Dev Biol ; 10: 895521, 2022.
Article in English | MEDLINE | ID: mdl-35832791

ABSTRACT

Heme plays a central role in diverse, life-essential processes that range from ubiquitous, housekeeping pathways such as respiration, to highly cell-specific ones such as oxygen transport by hemoglobin. The regulation of heme synthesis and its utilization is highly regulated and cell-specific. In this review, we have attempted to describe how the heme synthesis machinery is regulated by mitochondrial homeostasis as a means of coupling heme synthesis to its utilization and to the metabolic requirements of the cell. We have focused on discussing the regulation of mitochondrial heme synthesis enzymes by housekeeping proteins, transport of heme intermediates, and regulation of heme synthesis by macromolecular complex formation and mitochondrial metabolism. Recently discovered mechanisms are discussed in the context of the model organisms in which they were identified, while more established work is discussed in light of technological advancements.

6.
J Biol Chem ; 297(2): 100972, 2021 08.
Article in English | MEDLINE | ID: mdl-34280433

ABSTRACT

Heme plays a critical role in catalyzing life-essential redox reactions in all cells, and its synthesis must be tightly balanced with cellular requirements. Heme synthesis in eukaryotes is tightly regulated by the mitochondrial AAA+ unfoldase CLPX (caseinolytic mitochondrial matrix peptidase chaperone subunit X), which promotes heme synthesis by activation of δ-aminolevulinate synthase (ALAS/Hem1) in yeast and regulates turnover of ALAS1 in human cells. However, the specific mechanisms by which CLPX regulates heme synthesis are unclear. In this study, we interrogated the mechanisms by which CLPX regulates heme synthesis in erythroid cells. Quantitation of enzyme activity and protein degradation showed that ALAS2 stability and activity were both increased in the absence of CLPX, suggesting that CLPX primarily regulates ALAS2 by control of its turnover, rather than its activation. However, we also showed that CLPX is required for PPOX (protoporphyrinogen IX oxidase) activity and maintenance of FECH (ferrochelatase) levels, which are the terminal enzymes in heme synthesis, likely accounting for the heme deficiency and porphyrin accumulation observed in Clpx-/- cells. Lastly, CLPX is required for iron utilization for hemoglobin synthesis during erythroid differentiation. Collectively, our data show that the role of CLPX in yeast ALAS/Hem1 activation is not conserved in vertebrates as vertebrates rely on CLPX to regulate ALAS turnover as well as PPOX and FECH activity. Our studies reveal that CLPX mutations may cause anemia and porphyria via dysregulation of ALAS, FECH, and PPOX activities, as well as of iron metabolism.


Subject(s)
5-Aminolevulinate Synthetase/metabolism , Endopeptidase Clp/metabolism , Ferrochelatase/metabolism , Heme/biosynthesis , Iron/metabolism , Leukemia, Erythroblastic, Acute/pathology , Mitochondria/metabolism , Animals , Cell Line, Tumor , Endopeptidase Clp/genetics , Enzyme Activation , Gene Knockout Techniques/methods , Leukemia, Erythroblastic, Acute/enzymology , Leukemia, Erythroblastic, Acute/genetics , Mice , Models, Animal , Proteolysis , Zebrafish
8.
Development ; 148(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33318149

ABSTRACT

Mutations in the RNA helicase DDX3 have emerged as a frequent cause of intellectual disability in humans. Because many individuals carrying DDX3 mutations have additional defects in craniofacial structures and other tissues containing neural crest (NC)-derived cells, we hypothesized that DDX3 is also important for NC development. Using Xenopus tropicalis as a model, we show that DDX3 is required for normal NC induction and craniofacial morphogenesis by regulating AKT kinase activity. Depletion of DDX3 decreases AKT activity and AKT-dependent inhibitory phosphorylation of GSK3ß, leading to reduced levels of ß-catenin and Snai1: two GSK3ß substrates that are crucial for NC induction. DDX3 function in regulating these downstream signaling events during NC induction is likely mediated by RAC1, a small GTPase whose translation depends on the RNA helicase activity of DDX3. These results suggest an evolutionarily conserved role of DDX3 in NC development by promoting AKT activity, and provide a potential mechanism for the NC-related birth defects displayed by individuals harboring mutations in DDX3 and its downstream effectors in this signaling cascade.


Subject(s)
DEAD-box RNA Helicases/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Xenopus/metabolism , Animals , Cartilage/embryology , Cartilage/metabolism , Embryo, Nonmammalian/metabolism , Face/embryology , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3 beta/metabolism , Morphogenesis/genetics , Phosphorylation , Protein Stability , Skull/embryology , Skull/metabolism , Snail Family Transcription Factors/metabolism , Wnt Signaling Pathway , Xenopus/genetics , beta Catenin/metabolism , rac1 GTP-Binding Protein/metabolism
9.
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
10.
Proc Natl Acad Sci U S A ; 114(38): E8045-E8052, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28874591

ABSTRACT

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.


Subject(s)
5-Aminolevulinate Synthetase/metabolism , Endopeptidase Clp , Mutation, Missense , Porphyria, Erythropoietic , Protoporphyrins/biosynthesis , 5-Aminolevulinate Synthetase/genetics , Adolescent , Amino Acid Substitution , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism , Enzyme Stability/genetics , Female , Humans , Male , Porphyria, Erythropoietic/genetics , Porphyria, Erythropoietic/metabolism , Porphyria, Erythropoietic/pathology , Protoporphyrins/genetics
11.
J Biol Chem ; 292(39): 16284-16299, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28808058

ABSTRACT

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.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation, Enzymologic , Heme/biosynthesis , Zebrafish Proteins/metabolism , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Amino Acid Motifs , Amino Acid Substitution , Animals , Basic-Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic-Leucine Zipper Transcription Factors/genetics , Embryo, Nonmammalian/enzymology , Embryo, Nonmammalian/metabolism , Fanconi Anemia Complementation Group Proteins , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Microinjections , Morpholinos/metabolism , Mutation , RNA Interference , RNA, Small Interfering , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
12.
Science ; 356(6338): 608-616, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28495746

ABSTRACT

Multiple human diseases ensue from a hereditary or acquired deficiency of iron-transporting protein function that diminishes transmembrane iron flux in distinct sites and directions. Because other iron-transport proteins remain active, labile iron gradients build up across the corresponding protein-deficient membranes. Here we report that a small-molecule natural product, hinokitiol, can harness such gradients to restore iron transport into, within, and/or out of cells. The same compound promotes gut iron absorption in DMT1-deficient rats and ferroportin-deficient mice, as well as hemoglobinization in DMT1- and mitoferrin-deficient zebrafish. These findings illuminate a general mechanistic framework for small molecule-mediated site- and direction-selective restoration of iron transport. They also suggest that small molecules that partially mimic the function of missing protein transporters of iron, and possibly other ions, may have potential in treating human diseases.


Subject(s)
Iron/metabolism , Animals , Caco-2 Cells , Gastrointestinal Absorption , Hemoglobins/metabolism , Humans , Iron-Binding Proteins/metabolism , Monoterpenes/metabolism , Rats , Saccharomyces cerevisiae/metabolism , Tropolone/analogs & derivatives , Tropolone/metabolism
15.
Oncotarget ; 6(25): 20742-3, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26369700

ABSTRACT

Comment on: Yien Y, et al. TMEM14C is required for erythroid mitochondrial heme metabolism. J. Clin. Invest. 2014; 124:4294-4304.


Subject(s)
Erythroid Cells/metabolism , Mitochondria/metabolism , Protoporphyrins/physiology , Animals , Biological Transport , Heme/chemistry , Homeostasis , Humans , Mice , Sequence Analysis, RNA , Zebrafish
16.
Cell ; 161(4): 858-67, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25957689

ABSTRACT

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.


Subject(s)
Endopeptidase Clp/metabolism , Erythropoiesis , Eukaryota/metabolism , Heme/biosynthesis , 5-Aminolevulinate Synthetase/metabolism , Amino Acid Sequence , Aminolevulinic Acid/metabolism , Animals , Biological Evolution , Endopeptidase Clp/chemistry , Endopeptidase Clp/genetics , Eukaryota/genetics , Humans , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Molecular Chaperones/metabolism , Molecular Sequence Data , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Sequence Alignment , Zebrafish/metabolism
17.
Sci Signal ; 8(372): ra34, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25872869

ABSTRACT

In multicellular organisms, the mechanisms by which diverse cell types acquire distinct amino acids and how cellular function adapts to their availability are fundamental questions in biology. We found that increased neutral essential amino acid (NEAA) uptake was a critical component of erythropoiesis. As red blood cells matured, expression of the amino acid transporter gene Lat3 increased, which increased NEAA import. Inadequate NEAA uptake by pharmacologic inhibition or RNAi-mediated knockdown of LAT3 triggered a specific reduction in hemoglobin production in zebrafish embryos and murine erythroid cells through the mTORC1 (mammalian target of rapamycin complex 1)/4E-BP (eukaryotic translation initiation factor 4E-binding protein) pathway. CRISPR-mediated deletion of members of the 4E-BP family in murine erythroid cells rendered them resistant to mTORC1 and LAT3 inhibition and restored hemoglobin production. These results identify a developmental role for LAT3 in red blood cells and demonstrate that mTORC1 serves as a homeostatic sensor that couples hemoglobin production at the translational level to sufficient uptake of NEAAs, particularly L-leucine.


Subject(s)
Carrier Proteins/metabolism , Eukaryotic Initiation Factors/metabolism , Hemoglobins/metabolism , Leucine/metabolism , Multiprotein Complexes/metabolism , Phosphoproteins/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Transport Systems, Basic/genetics , Amino Acid Transport Systems, Basic/metabolism , Animals , Animals, Genetically Modified , CRISPR-Cas Systems , Carrier Proteins/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cells, Cultured , Embryo, Mammalian/blood supply , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Erythroid Cells/metabolism , Erythropoiesis/genetics , Eukaryotic Initiation Factors/genetics , Gene Expression Regulation, Developmental , HEK293 Cells , Hemoglobins/genetics , Humans , Immunoblotting , Mechanistic Target of Rapamycin Complex 1 , Mice , Microscopy, Confocal , Multiprotein Complexes/genetics , Phosphoproteins/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , Zebrafish
18.
Exp Hematol ; 43(1): 65-70, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25283745

ABSTRACT

Alternative splicing has emerged as a vital way to expand the functional repertoire of a set number of mammalian genes. For example, such changes can dramatically alter the function and cellular localization of transcription factors. With this in mind, we addressed whether EKLF/KLF1 mRNA, coding for a transcription factor that plays a critical role in erythropoietic gene regulation, is alternatively spliced. We find that EKLF mRNA undergoes exon skipping only in primary tissues and that this splice variant (SV) remains at a very low level in both embryonic and adult erythroid cells, as well as during terminal differentiation. The resultant protein is truncated and partially encodes a non-erythroid Krüppel-like factor amino acid sequence. Its overexpression can alter full-length erythroid Krüppel-like factor function at selected promoters. We discuss these results in the context of stress and with respect to recent global studies on the role of alternative splicing during terminal erythroid differentiation.


Subject(s)
Alternative Splicing , Erythroid Cells/metabolism , Erythropoiesis/genetics , Kruppel-Like Transcription Factors/genetics , Amino Acid Motifs , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , Cell Lineage , Female , Gene Expression Regulation , Genes, Reporter , Humans , K562 Cells , Kruppel-Like Transcription Factors/physiology , Leukemia, Erythroblastic, Acute/pathology , Mice , Molecular Sequence Data , Phlebotomy , Promoter Regions, Genetic , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Structure, Tertiary , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Recombinant Fusion Proteins/metabolism , Spleen/metabolism , Transcription, Genetic , Transcriptional Activation
19.
J Clin Invest ; 124(10): 4294-304, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25157825

ABSTRACT

The transport and intracellular trafficking of heme biosynthesis intermediates are crucial for hemoglobin production, which is a critical process in developing red cells. Here, we profiled gene expression in terminally differentiating murine fetal liver-derived erythroid cells to identify regulators of heme metabolism. We determined that TMEM14C, an inner mitochondrial membrane protein that is enriched in vertebrate hematopoietic tissues, is essential for erythropoiesis and heme synthesis in vivo and in cultured erythroid cells. In mice, TMEM14C deficiency resulted in porphyrin accumulation in the fetal liver, erythroid maturation arrest, and embryonic lethality due to profound anemia. Protoporphyrin IX synthesis in TMEM14C-deficient erythroid cells was blocked, leading to an accumulation of porphyrin precursors. The heme synthesis defect in TMEM14C-deficient cells was ameliorated with a protoporphyrin IX analog, indicating that TMEM14C primarily functions in the terminal steps of the heme synthesis pathway. Together, our data demonstrate that TMEM14C facilitates the import of protoporphyrinogen IX into the mitochondrial matrix for heme synthesis and subsequent hemoglobin production. Furthermore, the identification of TMEM14C as a protoporphyrinogen IX importer provides a genetic tool for further exploring erythropoiesis and congenital anemias.


Subject(s)
Erythropoiesis/genetics , Heme/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Anemia/metabolism , Animals , Cell Line , Erythroid Cells/metabolism , Gene Expression Regulation , Hemoglobins/metabolism , Liver/embryology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membranes/metabolism , Porphyrins/metabolism , Protoporphyrins/metabolism , RNA, Small Interfering/metabolism
20.
J Biol Chem ; 289(31): 21312-24, 2014 08 01.
Article in English | MEDLINE | ID: mdl-24907273

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems/physiology , Gene Deletion , Interspersed Repetitive Sequences , Animals , Base Sequence , Cell Line, Tumor , Genomics , Mice , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL