Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 610(7933): 768-774, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36261532

RESUMO

Haem is an iron-containing tetrapyrrole that is critical for a variety of cellular and physiological processes1-3. Haem binding proteins are present in almost all cellular compartments, but the molecular mechanisms that regulate the transport and use of haem within the cell remain poorly understood2,3. Here we show that haem-responsive gene 9 (HRG-9) (also known as transport and Golgi organization 2 (TANGO2)) is an evolutionarily conserved haem chaperone with a crucial role in trafficking haem out of haem storage or synthesis sites in eukaryotic cells. Loss of Caenorhabditis elegans hrg-9 and its paralogue hrg-10 results in the accumulation of haem in lysosome-related organelles, the haem storage site in worms. Similarly, deletion of the hrg-9 homologue TANGO2 in yeast and mammalian cells induces haem overload in mitochondria, the site of haem synthesis. We demonstrate that TANGO2 binds haem and transfers it from cellular membranes to apo-haemoproteins. Notably, homozygous tango2-/- zebrafish larvae develop pleiotropic symptoms including encephalopathy, cardiac arrhythmia and myopathy, and die during early development. These defects partially resemble the symptoms of human TANGO2-related metabolic encephalopathy and arrhythmias, a hereditary disease caused by mutations in TANGO24-8. Thus, the identification of HRG-9 as an intracellular haem chaperone provides a biological basis for exploring the aetiology and treatment of TANGO2-related disorders.


Assuntos
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Heme , Animais , Humanos , Arritmias Cardíacas/metabolismo , Encefalopatias/metabolismo , Caenorhabditis elegans/citologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Heme/metabolismo , Mitocôndrias/metabolismo , Chaperonas Moleculares/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
2.
Cell ; 145(5): 720-31, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21620137

RESUMO

Extracellular free heme can intercalate into membranes and promote damage to cellular macromolecules. Thus it is likely that specific intercellular pathways exist for the directed transport, trafficking, and delivery of heme to cellular destinations, although none have been found to date. Here we show that Caenorhabditis elegans HRG-3 is required for the delivery of maternal heme to developing embryos. HRG-3 binds heme and is exclusively secreted by maternal intestinal cells into the interstitial fluid for transport of heme to extraintestinal cells, including oocytes. HRG-3 deficiency results either in death during embryogenesis or in developmental arrest immediately post-hatching-phenotypes that are fully suppressed by maternal but not zygotic hrg-3 expression. Our results establish a role for HRG-3 as an intercellular heme-trafficking protein.


Assuntos
Caenorhabditis elegans/embriologia , Caenorhabditis elegans/metabolismo , Heme/metabolismo , Hemeproteínas/metabolismo , Animais , Animais Geneticamente Modificados , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Heme/deficiência , Hemeproteínas/química , Hemeproteínas/genética , Mucosa Intestinal/metabolismo , Mutação , Fenótipo , Transporte Proteico , Via Secretória
3.
Blood ; 140(10): 1145-1155, 2022 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-35820059

RESUMO

Developing erythroblasts acquire massive amounts of iron through the transferrin (Tf) cycle, which involves endocytosis, sorting, and recycling of the Tf-Tf receptor (Tfrc) complex. Previous studies on the hemoglobin-deficit (hbd) mouse have shown that the exocyst complex is indispensable for the Tfrc recycling; however, the precise mechanism underlying the efficient exocytosis and recycling of Tfrc in erythroblasts remains unclear. Here, we identify the guanine nucleotide exchange factor Grab as a critical regulator of the Tf cycle and iron metabolism during erythropoiesis. Grab is highly expressed in differentiating erythroblasts. Loss of Grab diminishes the Tfrc recycling and iron uptake, leading to hemoglobinization defects in mouse primary erythroblasts, mammalian erythroleukemia cells, and zebrafish embryos. These defects can be alleviated by supplementing iron together with hinokitiol, a small-molecule natural compound that can mediate iron transport independent of the Tf cycle. Mechanistically, Grab regulates the exocytosis of Tfrc-associated vesicles by activating the GTPase Rab8, which subsequently promotes the recruitment of the exocyst complex and vesicle exocytosis. Our results reveal a critical role for Grab in regulating the Tf cycle and provide new insights into iron homeostasis and erythropoiesis.


Assuntos
Eritroblastos , Fatores de Troca do Nucleotídeo Guanina , Ferro , Receptores da Transferrina , Animais , Eritroblastos/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Ferro/metabolismo , Mamíferos/metabolismo , Camundongos , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Transferrina/metabolismo , Peixe-Zebra/metabolismo
4.
FASEB J ; 37(2): e22757, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36607310

RESUMO

Vesicle trafficking is a fundamental cellular process that controls the transport of various proteins and cargos between cellular compartments in eukaryotes. Using a combination of genome-wide CRISPR screening in mammalian cells and RNAi screening in Caenorhabditis elegans, we identify chaperonin containing TCP-1 subunit 4 (CCT4) as a critical regulator of protein secretion and vesicle trafficking. In C. elegans, deficiency of cct-4 as well as other CCT subunits impairs the trafficking of endocytic markers in intestinal cells, and this defect resembles that of dyn-1 RNAi worms. Consistent with these findings, the silencing of CCT4 in human cells leads to defective endosomal trafficking, and this defect can be rescued by the dynamin activator Ryngo 1-23. These results suggest that the cytosolic chaperonin CCT may regulate vesicle trafficking by promoting the folding of dynamin in addition to its known substrate tubulin. Our findings establish an essential role for the CCT chaperonin in regulating vesicle trafficking, and provide new insights into the regulation of vesicle trafficking and the cellular function of the cytosolic chaperonin.


Assuntos
Caenorhabditis elegans , Chaperonina com TCP-1 , Animais , Humanos , Chaperonina com TCP-1/genética , Chaperonina com TCP-1/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Chaperoninas/genética , Chaperoninas/metabolismo , Tubulina (Proteína)/metabolismo , Citosol/metabolismo , Dobramento de Proteína , Mamíferos/metabolismo
5.
Blood ; 135(3): 208-219, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31945154

RESUMO

Mammalian red blood cells lack nuclei. The molecular mechanisms underlying erythroblast nuclear condensation and enucleation, however, remain poorly understood. Here we show that Wdr26, a gene upregulated during terminal erythropoiesis, plays an essential role in regulating nuclear condensation in differentiating erythroblasts. Loss of Wdr26 induces anemia in zebrafish and enucleation defects in mouse erythroblasts because of impaired erythroblast nuclear condensation. As part of the glucose-induced degradation-deficient ubiquitin ligase complex, Wdr26 regulates the ubiquitination and degradation of nuclear proteins, including lamin B. Failure of lamin B degradation blocks nuclear opening formation leading to impaired clearance of nuclear proteins and delayed nuclear condensation. Collectively, our study reveals an unprecedented role of an E3 ubiquitin ligase in regulating nuclear condensation and enucleation during terminal erythropoiesis. Our results provide mechanistic insights into nuclear protein homeostasis and vertebrate red blood cell development.


Assuntos
Diferenciação Celular , Núcleo Celular/metabolismo , Eritroblastos/fisiologia , Eritropoese , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Núcleo Celular/genética , Eritroblastos/citologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Proteínas de Peixe-Zebra/genética
6.
Int J Mol Sci ; 22(19)2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34638816

RESUMO

Vertebrates generate mature red blood cells (RBCs) via a highly regulated, multistep process called erythropoiesis. Erythropoiesis involves synthesis of heme and hemoglobin, clearance of the nuclei and other organelles, and remodeling of the plasma membrane, and these processes are exquisitely coordinated by specific regulatory factors including transcriptional factors and signaling molecules. Defects in erythropoiesis can lead to blood disorders such as congenital dyserythropoietic anemias, Diamond-Blackfan anemias, sideroblastic anemias, myelodysplastic syndrome, and porphyria. The molecular mechanisms of erythropoiesis are highly conserved between fish and mammals, and the zebrafish (Danio rerio) has provided a powerful genetic model for studying erythropoiesis. Studies in zebrafish have yielded important insights into RBC development and established a number of models for human blood diseases. Here, we focus on latest discoveries of the molecular processes and mechanisms regulating zebrafish erythropoiesis and summarize newly established zebrafish models of human anemias.


Assuntos
Anemia Aplástica , Desenvolvimento Embrionário , Eritrócitos/metabolismo , Eritropoese , Peixe-Zebra/embriologia , Anemia Aplástica/genética , Anemia Aplástica/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Peixe-Zebra/genética
7.
PLoS Genet ; 13(7): e1006892, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28692648

RESUMO

Recent studies found that mutations in the human SLC30A10 gene, which encodes a manganese (Mn) efflux transporter, are associated with hypermanganesemia with dystonia, polycythemia, and cirrhosis (HMDPC). However, the relationship between Mn metabolism and HMDPC is poorly understood, and no specific treatments are available for this disorder. Here, we generated two zebrafish slc30a10 mutant lines using the CRISPR/Cas9 system. Compared to wild-type animals, mutant adult animals developed significantly higher systemic Mn levels, and Mn accumulated in the brain and liver of mutant embryos in response to exogenous Mn. Interestingly, slc30a10 mutants developed neurological deficits in adulthood, as well as environmental Mn-induced manganism in the embryonic stage; moreover, mutant animals had impaired dopaminergic and GABAergic signaling. Finally, mutant animals developed steatosis, liver fibrosis, and polycythemia accompanied by increased epo expression. This phenotype was rescued partially by EDTA- CaNa2 chelation therapy and iron supplementation. Interestingly, prior to the onset of slc30a10 expression, expressing ATP2C1 (ATPase secretory pathway Ca2+ transporting 1) protected mutant embryos from Mn exposure, suggesting a compensatory role for Atp2c1 in the absence of Slc30a10. Notably, expressing either wild-type or mutant forms of SLC30A10 was sufficient to inhibit the effect of ATP2C1 in response to Mn challenge in both zebrafish embryos and HeLa cells. These findings suggest that either activating ATP2C1 or restoring the Mn-induced trafficking of ATP2C1 can reduce Mn accumulation, providing a possible target for treating HMDPC.


Assuntos
ATPases Transportadoras de Cálcio/genética , Proteínas de Transporte de Cátions/genética , Homeostase/genética , Manganês/metabolismo , Doenças Metabólicas/genética , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Sistemas CRISPR-Cas , Proteínas de Transporte de Cátions/deficiência , Genótipo , Células HeLa , Humanos , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Mutação , Peixe-Zebra/genética , Transportador 8 de Zinco
8.
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
9.
Nature ; 491(7425): 608-12, 2012 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-23135403

RESUMO

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.


Assuntos
Eritroblastos/metabolismo , Eritropoese , Heme/biossíntese , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas/metabolismo , Anemia Sideroblástica/genética , Anemia Sideroblástica/metabolismo , Anemia Sideroblástica/patologia , Animais , Modelos Animais de Doenças , Eritroblastos/citologia , Ferroquelatase/metabolismo , Teste de Complementação Genética , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Mitocôndrias/patologia , Proteínas Mitocondriais/deficiência , Proteínas Mitocondriais/genética , Oxirredução , Proteínas/genética , Peixe-Zebra/metabolismo , Proteína Inibidora de ATPase
10.
Pflugers Arch ; 466(8): 1605-18, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24668450

RESUMO

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.


Assuntos
Antiportadores de Cloreto-Bicarbonato/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Sequência de Aminoácidos , Animais , Antiportadores de Cloreto-Bicarbonato/metabolismo , Clonagem Molecular , Humanos , Hibridização In Situ , Microscopia Confocal , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
11.
J Biol Chem ; 287(12): 9601-12, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22303006

RESUMO

The roundworm Caenorhabditis elegans is a heme auxotroph that requires the coordinated actions of HRG-1 heme permeases to transport environmental heme into the intestine and HRG-3, a secreted protein, to deliver intestinal heme to other tissues including the embryo. Here we show that heme homeostasis in the extraintestinal hypodermal tissue was facilitated by the transmembrane protein HRG-2. Systemic heme deficiency up-regulated hrg-2 mRNA expression over 200-fold in the main body hypodermal syncytium, hyp 7. HRG-2 is a type I membrane protein that binds heme and localizes to the endoplasmic reticulum and apical plasma membrane. Cytochrome heme profiles are aberrant in HRG-2-deficient worms, a phenotype that was partially suppressed by heme supplementation. A heme-deficient yeast strain, ectopically expressing worm HRG-2, revealed significantly improved growth at submicromolar concentrations of exogenous heme. Taken together, our results implicate HRG-2 as a facilitator of heme utilization in the Caenorhabditis elegans hypodermis and provide a mechanism for the regulation of heme homeostasis in an extraintestinal tissue.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Heme/metabolismo , Hemeproteínas/metabolismo , Tela Subcutânea/metabolismo , Sequência de Aminoácidos , Animais , Caenorhabditis elegans/química , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/genética , Linhagem Celular , Hemeproteínas/química , Hemeproteínas/genética , Humanos , Dados de Sequência Molecular , Alinhamento de Sequência
12.
Biochim Biophys Acta ; 1823(9): 1459-67, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22285816

RESUMO

Iron plays an essential role in cellular metabolism and biological processes. However, due to its intrinsic redox activity, free iron is a potentially toxic molecule in cellular biochemistry. Thus, organisms have developed sophisticated ways to import, sequester, and utilize iron. The transferrin cycle is a well-studied iron uptake pathway that is important for most vertebrate cells. Circulating iron can also be imported into cells by mechanisms that are independent of transferrin. Once imported into erythroid cells, iron is predominantly consumed by the mitochondria for the biosynthesis of heme and iron sulfur clusters. This review focuses on canonical transferrin-mediated and the newly discovered, non-transferrin mediated iron uptake pathways, as well as, mitochondrial iron homeostasis in higher eukaryotes. This article is part of a Special Issue entitled: Cell Biology of Metals.


Assuntos
Células Eucarióticas/metabolismo , Ferro/metabolismo , Mitocôndrias/metabolismo , Receptores da Transferrina/metabolismo , Transferrina/metabolismo , Animais , Transporte Biológico/fisiologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Endossomos/metabolismo , Regulação da Expressão Gênica , Homeostase/fisiologia , Humanos , Deficiências de Ferro , Proteínas Reguladoras de Ferro/genética , Proteínas Reguladoras de Ferro/metabolismo , Receptores da Transferrina/genética , Sideróforos/metabolismo , Transferrina/genética , Vertebrados
13.
Curr Opin Hematol ; 19(3): 156-62, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22406824

RESUMO

PURPOSE OF REVIEW: Heme biosynthesis requires a series of enzymatic reactions that take place in the cytosol and the mitochondria as well as the proper intercellular and intracellular trafficking of iron. Heme can also be acquired by intestinal absorption and intercellular transport. The purpose of this review is to highlight recent work on heme and iron transport with an emphasis on their relevance in erythropoiesis. RECENT FINDINGS: Whereas the enzymes responsible for heme biosynthesis have been identified, transport mechanisms for iron, heme, or heme synthesis intermediates are only emerging. Recent studies have shed light on how these molecules are transported among various cellular compartments, as well as tissues. Much of this progress can be attributed to the use of model organisms such as S. cerevisiae, C. elegans, D. rerio, and M. musculus. Genetic studies in these models have led to the identification of several new genes involved in heme metabolism. Although our understanding has greatly improved, it is highly likely that other regulators exist and additional work is required to characterize the pathways by which heme and iron are transported within the erythron. SUMMARY: The identification of heme and iron transport mechanisms will improve our understanding of blood development and provide new insight into human blood disorders.


Assuntos
Eritropoese/fisiologia , Heme/biossíntese , Ferro/metabolismo , Animais , Transporte Biológico/fisiologia , Heme/fisiologia , Humanos , Mitocôndrias/metabolismo , Porfirinas/metabolismo
14.
Biomolecules ; 13(7)2023 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-37509184

RESUMO

Heme is an iron-containing tetrapyrrole that plays a critical role in various biological processes, including oxygen transport, electron transport, signal transduction, and catalysis. However, free heme is hydrophobic and potentially toxic to cells. Organisms have evolved specific pathways to safely transport this essential but toxic macrocycle within and between cells. The bacterivorous soil-dwelling nematode Caenorhabditis elegans is a powerful animal model for studying heme-trafficking pathways, as it lacks the ability to synthesize heme but instead relies on specialized trafficking pathways to acquire, distribute, and utilize heme. Over the past 15 years, studies on this microscopic animal have led to the identification of a number of heme-trafficking proteins, with corresponding functional homologs in vertebrates. In this review, we provide a comprehensive overview of the heme-trafficking proteins identified in C. elegans and their corresponding homologs in related organisms.


Assuntos
Caenorhabditis elegans , Nematoides , Animais , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Heme/metabolismo , Homeostase/fisiologia , Nematoides/metabolismo , Transporte Biológico/fisiologia
15.
J Biol Chem ; 285(35): 26753-26759, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20522548

RESUMO

Iron is an essential element for diverse biological functions. In mammals, the majority of iron is enclosed within a single prosthetic group: heme. In metazoans, heme is synthesized via a highly conserved and coordinated pathway within the mitochondria. However, iron is acquired from the environment and subsequently assimilated into various cellular pathways, including heme synthesis. Both iron and heme are toxic but essential cofactors. How is iron transported from the extracellular milieu to the mitochondria? How are heme and heme intermediates coordinated with iron transport? Although recent studies have answered some questions, several pieces of this intriguing puzzle remain unsolved.


Assuntos
Heme/biossíntese , Ferro/metabolismo , Mitocôndrias/metabolismo , Porfirinas/metabolismo , Animais , Transporte Biológico/fisiologia , Humanos
16.
J Genet Genomics ; 47(11): 694-704, 2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33547005

RESUMO

Mitochondria are the central hub for many metabolic processes, including the citric acid cycle, oxidative phosphorylation, and fatty acid oxidation. Recent studies have identified a new mitochondrial protein family, Fam210, that regulates bone metabolism and red cell development in vertebrates. The model organism Caenorhabditis elegans has a Fam210 gene, y56a3a.22, but it lacks both bones and red blood cells. In this study, we report that Y56A3A.22 plays a crucial role in regulating mitochondrial protein homeostasis and reproduction. The nematode y56a3a.22 is expressed in various tissues, including the intestine, muscle, hypodermis, and germline, and its encoded protein is predominantly localized in mitochondria. y56a3a.22 deletion mutants are sterile owing to impaired oogenesis. Loss of Y56A3A.22 induced mitochondrial unfolded protein response (UPRmt), which is mediated through the ATFS-1-dependent pathway, in tissues such as the intestine, germline, hypodermis, and vulval muscle. We further show that infertility and UPRmt induces by Y56A3A.22 deficiency are not attributed to systemic iron deficiency. Together, our study reveals an important role of Y56A3A.22 in regulating mitochondrial protein homeostasis and oogenesis and provides a new genetic tool for exploring the mechanisms regulating mitochondrial metabolism and reproduction as well as the fundamental role of the Fam210 family.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Mitocôndrias/genética , Oogênese/genética , Fatores de Transcrição/genética , Resposta a Proteínas não Dobradas/genética , Animais , Osso e Ossos/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/crescimento & desenvolvimento , Eritrócitos/metabolismo , Feminino , Células Germinativas/crescimento & desenvolvimento , Células Germinativas/metabolismo , Humanos , Intestinos/crescimento & desenvolvimento , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Reprodução/genética , Transdução de Sinais/genética , Vulva/crescimento & desenvolvimento , Vulva/metabolismo
17.
J Clin Invest ; 130(10): 5245-5256, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32634119

RESUMO

The congenital sideroblastic anemias (CSAs) can be caused by primary defects in mitochondrial iron-sulfur (Fe-S) cluster biogenesis. HSCB (heat shock cognate B), which encodes a mitochondrial cochaperone, also known as HSC20 (heat shock cognate protein 20), is the partner of mitochondrial heat shock protein A9 (HSPA9). Together with glutaredoxin 5 (GLRX5), HSCB and HSPA9 facilitate the transfer of nascent 2-iron, 2-sulfur clusters to recipient mitochondrial proteins. Mutations in both HSPA9 and GLRX5 have previously been associated with CSA. Therefore, we hypothesized that mutations in HSCB could also cause CSA. We screened patients with genetically undefined CSA and identified a frameshift mutation and a rare promoter variant in HSCB in a female patient with non-syndromic CSA. We found that HSCB expression was decreased in patient-derived fibroblasts and K562 erythroleukemia cells engineered to have the patient-specific promoter variant. Furthermore, gene knockdown and deletion experiments performed in K562 cells, zebrafish, and mice demonstrate that loss of HSCB results in impaired Fe-S cluster biogenesis, a defect in RBC hemoglobinization, and the development of siderocytes and more broadly perturbs hematopoiesis in vivo. These results further affirm the involvement of Fe-S cluster biogenesis in erythropoiesis and hematopoiesis and define HSCB as a CSA gene.


Assuntos
Anemia Sideroblástica/genética , Chaperonas Moleculares/genética , Mutação , Adolescente , Anemia Sideroblástica/congênito , Anemia Sideroblástica/metabolismo , Animais , Criança , Análise Mutacional de DNA , Feminino , Mutação da Fase de Leitura , Técnicas de Silenciamento de Genes , Humanos , Proteínas Ferro-Enxofre/deficiência , Proteínas Ferro-Enxofre/genética , Células K562 , Masculino , Camundongos , Camundongos Knockout , Chaperonas Moleculares/metabolismo , Linhagem , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Adulto Jovem , Peixe-Zebra
18.
Sci China Life Sci ; 58(8): 757-64, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26100009

RESUMO

Heme is an iron-containing tetrapyrrole that plays a critical role in regulating a variety of biological processes including oxygen and electron transport, gas sensing, signal transduction, biological clock, and microRNA processing. Most metazoan cells synthesize heme via a conserved pathway comprised of eight enzyme-catalyzed reactions. Heme can also be acquired from food or extracellular environment. Cellular heme homeostasis is maintained through the coordinated regulation of synthesis, transport, and degradation. This review presents the current knowledge of the synthesis and transport of heme in metazoans and highlights recent advances in the regulation of these pathways.


Assuntos
Vias Biossintéticas , Heme/biossíntese , Homeostase/fisiologia , Transdução de Sinais/fisiologia , 5-Aminolevulinato Sintetase/metabolismo , Animais , Transporte Biológico/fisiologia , Ferroquelatase/metabolismo , Ferro/metabolismo , Oxigênio/metabolismo
19.
Antioxid Redox Signal ; 22(15): 1325-36, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25608116

RESUMO

AIMS: Hereditary hemochromatosis (HH) is an iron overload disease that is caused by mutations in HFE, HJV, and several other genes. However, whether HFE-HH and HJV-HH share a common pathway via hepcidin regulation is currently unclear. Recently, some HH patients have been reported to carry concurrent mutations in both the HFE and HJV genes. To dissect the roles and molecular mechanisms of HFE and/or HJV in the pathogenesis of HH, we studied Hfe(-/-), Hjv(-/-), and Hfe(-/-)Hjv(-/-) double-knockout mouse models. RESULTS: Hfe(-/-)Hjv(-/-) mice developed iron overload in multiple organs at levels comparable to Hjv(-/-) mice. After an acute delivery of iron, the expression of hepcidin (i.e., Hamp1 mRNA) was increased in the livers of wild-type and Hfe(-/-) mice, but not in either Hjv(-/-) or Hfe(-/-)Hjv(-/-) mice. Furthermore, iron-induced phosphorylation of Smad1/5/8 was not detected in the livers of Hjv(-/-) or Hfe(-/-)Hjv(-/-) mice. INNOVATION: We generated and phenotypically characterized Hfe(-/-)Hjv(-/-) double-knockout mice. In addition, because they faithfully phenocopy clinical HH patients, these mouse models are an invaluable tool for mechanistically dissecting how HFE and HJV regulate hepcidin expression. CONCLUSIONS: Based on our results, we conclude that HFE may depend on HJV for transferrin-dependent hepcidin regulation. The presence of residual hepcidin in the absence of HFE suggests either the presence of an unknown regulator (e.g., TFR2) that is synergistic with HJV or that HJV is sufficient to maintain basal levels of hepcidin.


Assuntos
Hemocromatose/patologia , Hepcidinas/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Ferro/administração & dosagem , Proteínas de Membrana/genética , Animais , Modelos Animais de Doenças , Feminino , Proteínas Ligadas por GPI , Regulação da Expressão Gênica , Hemocromatose/genética , Hemocromatose/metabolismo , Proteína da Hemocromatose , Hepcidinas/genética , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Ferro/metabolismo , Fígado/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Transdução de Sinais
20.
Sci Signal ; 8(372): ra34, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25872869

RESUMO

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.


Assuntos
Proteínas de Transporte/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Hemoglobinas/metabolismo , Leucina/metabolismo , Complexos Multiproteicos/metabolismo , Fosfoproteínas/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sistemas de Transporte de Aminoácidos Básicos/genética , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Animais , Animais Geneticamente Modificados , Sistemas CRISPR-Cas , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Células Cultivadas , Embrião de Mamíferos/irrigação sanguínea , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Embrião não Mamífero/embriologia , Embrião não Mamífero/metabolismo , Células Eritroides/metabolismo , Eritropoese/genética , Fatores de Iniciação em Eucariotos/genética , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Hemoglobinas/genética , Humanos , Immunoblotting , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Microscopia Confocal , Complexos Multiproteicos/genética , Fosfoproteínas/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA