Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.114
Filtrar
1.
Int J Mol Sci ; 25(10)2024 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-38791308

RESUMEN

Heme biosynthesis is a highly conserved pathway from bacteria to higher animals. Heme, which serves as a prosthetic group for various enzymes involved in multiple biochemical processes, is essential in almost all species, making heme homeostasis vital for life. However, studies on the biological functions of heme in filamentous fungi are scarce. In this study, we investigated the role of heme in Fusarium graminearum. A mutant lacking the rate-limiting enzymes in heme synthesis, coproporphyrinogen III oxidase (Cpo) or ferrochelatase (Fc), was constructed using a homologous recombination strategy. The results showed that the absence of these enzymes was lethal to F. graminearum, but the growth defect could be rescued by the addition of hemin, so we carried out further studies with the help of hemin. The results demonstrated that heme was required for the activity of FgCyp51, and its absence increased the sensitivity to tebuconazole and led to the upregulation of FgCYP51 in F. graminearum. Additionally, heme plays an indispensable role in the life cycle of F. graminearum, which is essential for vegetative growth, conidiation, external stress response (especially oxidative stress), lipid accumulation, fatty acid ß-oxidation, autophagy, and virulence.


Asunto(s)
Fusarium , Hemo , Fusarium/efectos de los fármacos , Fusarium/metabolismo , Fusarium/crecimiento & desarrollo , Fusarium/genética , Hemo/biosíntesis , Hemo/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Estrés Fisiológico , Estrés Oxidativo/efectos de los fármacos , Triazoles/farmacología , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Fungicidas Industriales/farmacología , Ferroquelatasa/metabolismo , Ferroquelatasa/genética
2.
Mol Biol (Mosk) ; 57(6): 1085-1097, 2023.
Artículo en Ruso | MEDLINE | ID: mdl-38062963

RESUMEN

δ-Aminolevulinic acid dehydratase (ALAD) is a key enzyme of the cytoplasmic heme biosynthesis pathway. The primary structure of the ALAD gene, the multimeric structure of the ALAD/hemB protein, and ALAD expression during the annual reproductive cycle were studied in the cold-water marine sponge Halisarca dujardinii. The results implicated the GATA-1 transcription factor and DNA methylation in regulating ALAD expression. Re-aggregation of sponge cells was accompanied by a decrease in ALAD expression and a change in the cell content of an active ALAD/hemB form. Further study of heme biosynthesis and the role of ALAD/hemB in morphogenesis of basal animals may provide new opportunities for treating pathologies in higher animals.


Asunto(s)
Poríferos , Animales , Hemo/biosíntesis , Hemo/metabolismo , Poríferos/enzimología , Poríferos/metabolismo , Porfobilinógeno Sintasa/genética , Porfobilinógeno Sintasa/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(30): e2108245119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858410

RESUMEN

Heme is an oxygen carrier and a cofactor of both industrial enzymes and food additives. The intracellular level of free heme is low, which limits the synthesis of heme proteins. Therefore, increasing heme synthesis allows an increased production of heme proteins. Using the genome-scale metabolic model (GEM) Yeast8 for the yeast Saccharomyces cerevisiae, we identified fluxes potentially important to heme synthesis. With this model, in silico simulations highlighted 84 gene targets for balancing biomass and increasing heme production. Of those identified, 76 genes were individually deleted or overexpressed in experiments. Empirically, 40 genes individually increased heme production (up to threefold). Heme was increased by modifying target genes, which not only included the genes involved in heme biosynthesis, but also those involved in glycolysis, pyruvate, Fe-S clusters, glycine, and succinyl-coenzyme A (CoA) metabolism. Next, we developed an algorithmic method for predicting an optimal combination of these genes by using the enzyme-constrained extension of the Yeast8 model, ecYeast8. The computationally identified combination for enhanced heme production was evaluated using the heme ligand-binding biosensor (Heme-LBB). The positive targets were combined using CRISPR-Cas9 in the yeast strain (IMX581-HEM15-HEM14-HEM3-Δshm1-HEM2-Δhmx1-FET4-Δgcv2-HEM1-Δgcv1-HEM13), which produces 70-fold-higher levels of intracellular heme.


Asunto(s)
Hemo , Ingeniería Metabólica , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Simulación por Computador , Hemo/biosíntesis , Hemo/genética , Hemoproteínas/biosíntesis , Hemoproteínas/genética , Ingeniería Metabólica/métodos , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
4.
Sci Rep ; 12(1): 1472, 2022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35087136

RESUMEN

Obstructive sleep apnea (OSA) is a common disorder associated with increased risk of cardiovascular disease and mortality. Iron and heme metabolism, implicated in ventilatory control and OSA comorbidities, was associated with OSA phenotypes in recent admixture mapping and gene enrichment analyses. However, its causal contribution was unclear. In this study, we performed pathway-level transcriptional Mendelian randomization (MR) analysis to investigate the causal relationships between iron and heme related pathways and OSA. In primary analysis, we examined the expression level of four iron/heme Reactome pathways as exposures and four OSA traits as outcomes using cross-tissue cis-eQTLs from the Genotype-Tissue Expression portal and published genome-wide summary statistics of OSA. We identify a significant putative causal association between up-regulated heme biosynthesis pathway with higher sleep time percentage of hypoxemia (p = 6.14 × 10-3). This association is supported by consistency of point estimates in one-sample MR in the Multi-Ethnic Study of Atherosclerosis using high coverage DNA and RNA sequencing data generated by the Trans-Omics for Precision Medicine project. Secondary analysis for 37 additional iron/heme Gene Ontology pathways did not reveal any significant causal associations. This study suggests a causal association between increased heme biosynthesis and OSA severity.


Asunto(s)
Hemo/biosíntesis , Redes y Vías Metabólicas/genética , Apnea Obstructiva del Sueño/epidemiología , Anciano , Conjuntos de Datos como Asunto , Femenino , Predisposición Genética a la Enfermedad , Humanos , Hierro/metabolismo , Masculino , Análisis de la Aleatorización Mendeliana , Persona de Mediana Edad , Polisomnografía , Sitios de Carácter Cuantitativo , Índice de Severidad de la Enfermedad , Apnea Obstructiva del Sueño/sangre , Apnea Obstructiva del Sueño/diagnóstico , Apnea Obstructiva del Sueño/genética , Regulación hacia Arriba
5.
Am J Pathol ; 192(1): 4-17, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34924168

RESUMEN

Metabolic heterogeneity or functional zonation is a key characteristic of the liver that allows different metabolic pathways to be spatially regulated within the hepatic system and together contribute to whole body homeostasis. These metabolic pathways are segregated along the portocentral axis of the liver lobule into three hepatic zones: periportal, intermediate or midzonal, and perivenous. The liver performs complementary or opposing metabolic functions within different hepatic zones while synergistic functions are regulated by overlapping zones, thereby maintaining the overall physiological stability. The Wnt/ß-catenin signaling pathway is well known for its role in liver growth, development, and regeneration. In addition, the Wnt/ß-catenin pathway plays a fundamental and dominant role in hepatic zonation and signals to orchestrate various functions of liver metabolism and pathophysiology. The ß-catenin protein is the central player in the Wnt/ß-catenin signaling cascade, and its activation is crucial for metabolic patterning of the liver. However, dysregulation of Wnt/ß-catenin signaling is also implicated in different liver pathologies, including those associated with metabolic syndrome. ß-Catenin is preferentially localized in the central region of the hepatic lobule surrounding the central vein and regulates multiple functions of this region. This review outlines the role of Wnt/ß-catenin signaling pathway in controlling the different metabolic processes surrounding the central vein and its relation to liver homeostasis and dysfunction.


Asunto(s)
Homeostasis , Hígado/metabolismo , Vía de Señalización Wnt , Animales , Regulación de la Expresión Génica , Hemo/biosíntesis , Humanos , Vía de Señalización Wnt/genética , Xenobióticos/metabolismo
6.
Biomolecules ; 11(12)2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34944556

RESUMEN

This study hypothesizes that bacteria inhabiting shale rock affect the content of the sedimentary cobalt protoporphyrin present in it and can use it as a precursor for heme synthesis. To verify this hypothesis, we conducted qualitative and quantitative comparative analyses of cobalt protoporphyrin as well as heme, and heme iron in shale rock that were (i) inhabited by bacteria in the field, (ii) treated with bacteria in the laboratory, and with (iii) bacterial culture on synthetic cobalt protoporphyrin. Additionally, we examined the above-mentioned samples for the presence of enzymes involved in the heme biosynthesis and uptake as well as hemoproteins. We found depletion of cobalt protoporphyrin and a much higher heme concentration in the shale rock inhabited by bacteria in the field as well as the shale rock treated with bacteria in the laboratory. Similarly, we observed the accumulation of protoporphyrin in bacterial cells grown on synthetic cobalt protoporphyrin. We detected numerous hemoproteins in metaproteome of bacteria inhabited shale rock in the field and in proteomes of bacteria inhabited shale rock and synthetic cobalt protoporhyrin in the laboratory, but none of them had all the enzymes involved in the heme biosynthesis. However, proteins responsible for heme uptake, ferrochelatase and sirohydrochlorin cobaltochelatase/sirohydrochlorin cobalt-lyase were detected in all studied samples.


Asunto(s)
Bacterias/crecimiento & desarrollo , Fósiles/microbiología , Sedimentos Geológicos/microbiología , Hemo/análisis , Protoporfirinas/análisis , Bacterias/metabolismo , Proteínas Bacterianas/metabolismo , Técnicas Bacteriológicas , Medios de Cultivo/química , Ferroquelatasa/metabolismo , Regulación Bacteriana de la Expresión Génica , Sedimentos Geológicos/química , Hemo/biosíntesis , Liasas/metabolismo , Proteómica , Protoporfirinas/biosíntesis
7.
Microbiology (Reading) ; 167(10)2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34661520

RESUMEN

Uroporphyrinogen III, the universal progenitor of macrocyclic, modified tetrapyrroles, is produced from aminolaevulinic acid (ALA) by a conserved pathway involving three enzymes: porphobilinogen synthase (PBGS), hydroxymethylbilane synthase (HmbS) and uroporphyrinogen III synthase (UroS). The gene encoding uroporphyrinogen III synthase has not yet been identified in Plasmodium falciparum, but it has been suggested that this activity is housed inside a bifunctional hybroxymethylbilane synthase (HmbS). Additionally, an unknown protein encoded by PF3D7_1247600 has also been predicted to possess UroS activity. In this study it is demonstrated that neither of these proteins possess UroS activity and the real UroS remains to be identified. This was demonstrated by the failure of codon-optimized genes to complement a defined Escherichia coli hemD- mutant (SASZ31) deficient in UroS activity. Furthermore, HPLC analysis of the oxidized reaction product from recombinant, purified P. falciparum HmbS showed that only uroporphyrin I could be detected (corresponding to hydroxymethylbilane production). No uroporphyrin III was detected, showing that P. falciparum HmbS does not have UroS activity and can only catalyze the formation of hydroxymethylbilane from porphobilinogen.


Asunto(s)
Hemo/biosíntesis , Hidroximetilbilano Sintasa/metabolismo , Plasmodium falciparum/enzimología , Vías Biosintéticas , Escherichia coli/genética , Prueba de Complementación Genética , Hidroximetilbilano Sintasa/genética , Mutación , Plasmodium falciparum/genética , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo , Uroporfirinógeno III Sintetasa/genética , Uroporfirinógeno III Sintetasa/metabolismo , Uroporfirinógenos/metabolismo
8.
Biomolecules ; 11(9)2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34572607

RESUMEN

In the traditional fermentative model yeast Saccharomyces cerevisiae, ScIxr1 is an HMGB (High Mobility Group box B) protein that has been considered as an important regulator of gene transcription in response to external changes like oxygen, carbon source, or nutrient availability. Kluyveromyces lactis is also a useful eukaryotic model, more similar to many human cells due to its respiratory metabolism. We cloned and functionally characterized by different methodologies KlIXR1, which encodes a protein with only 34.4% amino acid sequence similarity to ScIxr1. Our data indicate that both proteins share common functions, including their involvement in the response to hypoxia or oxidative stress induced by hydrogen peroxide or metal treatments, as well as in the control of key regulators for maintenance of the dNTP (deoxyribonucleotide triphosphate) pool and ribosome synthesis. KlIxr1 is able to bind specific regulatory DNA sequences in the promoter of its target genes, which are well conserved between S. cerevisiae and K. lactis. Oppositely, we found important differences between ScIrx1 and KlIxr1 affecting cellular responses to cisplatin or cycloheximide in these yeasts, which could be dependent on specific and non-conserved domains present in these two proteins.


Asunto(s)
Desoxirribonucleótidos/metabolismo , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Proteínas HMGB/metabolismo , Kluyveromyces/crecimiento & desarrollo , Kluyveromyces/genética , Secuencia de Bases , Cadmio/toxicidad , Carbono/farmacología , Ciclo Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Proteínas Fúngicas/química , Eliminación de Gen , Regulación Fúngica de la Expresión Génica/efectos de los fármacos , Proteínas HMGB/química , Hemo/biosíntesis , Peróxido de Hidrógeno/toxicidad , Kluyveromyces/efectos de los fármacos , Mutación/genética , Oxidación-Reducción/efectos de los fármacos , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica/efectos de los fármacos , Procesamiento Postranscripcional del ARN/efectos de los fármacos , ARN Ribosómico/genética , Ribosomas/efectos de los fármacos , Ribosomas/metabolismo , Saccharomyces cerevisiae/metabolismo
9.
Biochem J ; 478(17): 3239-3252, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34402499

RESUMEN

Ferrochelatase catalyzes the insertion of ferrous iron into a porphyrin macrocycle to produce the essential cofactor, heme. In humans this enzyme not only catalyzes the terminal step, but also serves a regulatory step in the heme synthesis pathway. Over a dozen crystal structures of human ferrochelatase have been solved and many variants have been characterized kinetically. In addition, hydrogen deuterium exchange, resonance Raman, molecular dynamics, and high level quantum mechanic studies have added to our understanding of the catalytic cycle of the enzyme. However, an understanding of how the metal ion is delivered and the specific role that active site residues play in catalysis remain open questions. Data are consistent with metal binding and insertion occurring from the side opposite from where pyrrole proton abstraction takes place. To better understand iron delivery and binding as well as the role of conserved residues in the active site, we have constructed and characterized a series of enzyme variants. Crystallographic studies as well as rescue and kinetic analysis of variants were performed. Data from these studies are consistent with the M76 residue playing a role in active site metal binding and formation of a weak iron protein ligand being necessary for product release. Additionally, structural data support a role for E343 in proton abstraction and product release in coordination with a peptide loop composed of Q302, S303 and K304 that act a metal sensor.


Asunto(s)
Dominio Catalítico/fisiología , Ferroquelatasa/química , Ferroquelatasa/metabolismo , Modelos Moleculares , Biocatálisis , Cristalización , Hemo/biosíntesis , Histidina/metabolismo , Humanos , Hierro/metabolismo , Cinética , Ligandos , Unión Proteica , Protones , Protoporfirinas/metabolismo
10.
Crit Rev Biochem Mol Biol ; 56(6): 640-668, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34428995

RESUMEN

Aerobic respiration is a key energy-producing pathway in many prokaryotes and virtually all eukaryotes. The final step of aerobic respiration is most commonly catalyzed by heme-copper oxidases embedded in the cytoplasmic or mitochondrial membrane. The majority of these terminal oxidases contain a prenylated heme (typically heme a or occasionally heme o) in the active site. In addition, many heme-copper oxidases, including mitochondrial cytochrome c oxidases, possess a second heme a cofactor. Despite the critical role of heme a in the electron transport chain, the details of the mechanism by which heme b, the prototypical cellular heme, is converted to heme o and then to heme a remain poorly understood. Recent structural investigations, however, have helped clarify some elements of heme a biosynthesis. In this review, we discuss the insight gained from these advances. In particular, we present a new structural model of heme o synthase (HOS) based on distance restraints from inferred coevolutionary relationships and refined by molecular dynamics simulations that are in good agreement with the experimentally determined structures of HOS homologs. We also analyze the two structures of heme a synthase (HAS) that have recently been solved by other groups. For both HOS and HAS, we discuss the proposed catalytic mechanisms and highlight how new insights into the heme-binding site locations shed light on previously obtained biochemical data. Finally, we explore the implications of the new structural data in the broader context of heme trafficking in the heme a biosynthetic pathway and heme-copper oxidase assembly.


Asunto(s)
Transferasas Alquil y Aril/metabolismo , Proteínas Bacterianas/metabolismo , Hemo/análogos & derivados , Animales , Archaea/metabolismo , Bacterias/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Eucariontes/metabolismo , Hemo/biosíntesis , Hemo/metabolismo , Humanos , Simulación de Dinámica Molecular , Conformación Proteica , Transporte de Proteínas
11.
J Biol Chem ; 297(2): 100972, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34280433

RESUMEN

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.


Asunto(s)
5-Aminolevulinato Sintetasa/metabolismo , Endopeptidasa Clp/metabolismo , Ferroquelatasa/metabolismo , Hemo/biosíntesis , Hierro/metabolismo , Leucemia Eritroblástica Aguda/patología , Mitocondrias/metabolismo , Animales , Línea Celular Tumoral , Endopeptidasa Clp/genética , Activación Enzimática , Técnicas de Inactivación de Genes/métodos , Leucemia Eritroblástica Aguda/enzimología , Leucemia Eritroblástica Aguda/genética , Ratones , Modelos Animales , Proteolisis , Pez Cebra
12.
Genome Med ; 13(1): 108, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34225776

RESUMEN

BACKGROUND: Candidemia is one of the most common nosocomial bloodstream infections in the United States, causing significant morbidity and mortality in hospitalized patients, but the breadth of the host response to Candida infections in human patients remains poorly defined. METHODS: In order to better define the host response to Candida infection at the transcriptional level, we performed RNA sequencing on serial peripheral blood samples from 48 hospitalized patients with blood cultures positive for Candida species and compared them to patients with other acute viral, bacterial, and non-infectious illnesses. Regularized multinomial regression was utilized to develop pathogen class-specific gene expression classifiers. RESULTS: Candidemia triggers a unique, robust, and conserved transcriptomic response in human hosts with 1641 genes differentially upregulated compared to healthy controls. Many of these genes corresponded to components of the immune response to fungal infection, heavily weighted toward neutrophil activation, heme biosynthesis, and T cell signaling. We developed pathogen class-specific classifiers from these unique signals capable of identifying and differentiating candidemia, viral, or bacterial infection across a variety of hosts with a high degree of accuracy (auROC 0.98 for candidemia, 0.99 for viral and bacterial infection). This classifier was validated on two separate human cohorts (auROC 0.88 for viral infection and 0.87 for bacterial infection in one cohort; auROC 0.97 in another cohort) and an in vitro model (auROC 0.94 for fungal infection, 0.96 for bacterial, and 0.90 for viral infection). CONCLUSIONS: Transcriptional analysis of circulating leukocytes in patients with acute Candida infections defines novel aspects of the breadth of the human immune response during candidemia and suggests promising diagnostic approaches for simultaneously differentiating multiple types of clinical illnesses in at-risk, acutely ill patients.


Asunto(s)
Candidemia/etiología , Candidemia/metabolismo , Susceptibilidad a Enfermedades , Hemo/biosíntesis , Interacciones Huésped-Patógeno/genética , Activación Neutrófila/genética , Transcriptoma , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores , Candidemia/diagnóstico , Candidemia/tratamiento farmacológico , Estudios de Casos y Controles , Biología Computacional/métodos , Bases de Datos Genéticas , Femenino , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno/inmunología , Humanos , Masculino , Persona de Mediana Edad , Activación Neutrófila/inmunología , Pronóstico , Curva ROC , Reproducibilidad de los Resultados , Factores de Riesgo , Índice de Severidad de la Enfermedad
13.
Int J Mol Sci ; 22(10)2021 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-34064887

RESUMEN

Bacterial response to nitric oxide (NO) is of major importance for bacterial survival. NO stress is a main actor of the eukaryotic immune response and several pathogenic bacteria have developed means for detoxification and repair of the damages caused by NO. However, bacterial mechanisms of NO resistance by Gram-positive bacteria are poorly described. In the opportunistic foodborne pathogen Bacillus cereus, genome sequence analyses did not identify homologs to known NO reductases and transcriptional regulators, such as NsrR, which orchestrate the response to NO of other pathogenic or non-pathogenic bacteria. Using a transcriptomic approach, we investigated the adaptation of B. cereus to NO stress. A cluster of 6 genes was identified to be strongly up-regulated in the early phase of the response. This cluster contains an iron-sulfur cluster repair enzyme, a nitrite reductase and three enzymes involved in siroheme biosynthesis. The expression pattern and close genetic localization suggest a functional link between these genes, which may play a pivotal role in the resistance of B. cereus to NO stress during infection.


Asunto(s)
Bacillus cereus/metabolismo , Proteínas Bacterianas/metabolismo , Hemo/análogos & derivados , Hierro/metabolismo , Óxido Nítrico/toxicidad , Nitrito Reductasas/metabolismo , Estrés Oxidativo , Bacillus cereus/efectos de los fármacos , Bacillus cereus/genética , Bacillus cereus/crecimiento & desarrollo , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Hemo/biosíntesis , Transcripción Genética
14.
Cell Rep ; 35(11): 109252, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34133926

RESUMEN

Heme is an iron-containing porphyrin of vital importance for cell energetic metabolism. High rates of heme synthesis are commonly observed in proliferating cells. Moreover, the cell-surface heme exporter feline leukemia virus subgroup C receptor 1a (FLVCR1a) is overexpressed in several tumor types. However, the reasons why heme synthesis and export are enhanced in highly proliferating cells remain unknown. Here, we illustrate a functional axis between heme synthesis and heme export: heme efflux through the plasma membrane sustains heme synthesis, and implementation of the two processes down-modulates the tricarboxylic acid (TCA) cycle flux and oxidative phosphorylation. Conversely, inhibition of heme export reduces heme synthesis and promotes the TCA cycle fueling and flux as well as oxidative phosphorylation. These data indicate that the heme synthesis-export system modulates the TCA cycle and oxidative metabolism and provide a mechanistic basis for the observation that both processes are enhanced in cells with high-energy demand.


Asunto(s)
Ciclo del Ácido Cítrico , Hemo/biosíntesis , Fosforilación Oxidativa , Animales , Transporte Biológico , Células CACO-2 , Hemo/metabolismo , Humanos , Proteínas de Transporte de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones SCID , Receptores Virales/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Sci Rep ; 11(1): 7457, 2021 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-33811225

RESUMEN

Isolation of bacterial small colony variants (SCVs) from clinical specimens is not uncommon and can fundamentally change the outcome of the associated infections. Bacterial SCVs often emerge with their normal colony phenotype (NCV) co-isolates in the same sample. The basis of SCV emergence in vivo is not well understood in Gram-negative bacteria. In this study, we interrogated the causal genetic lesions of SCV growth in three pairs of NCV and SCV co-isolates of Escherichia coli, Citrobacter freundii, and Enterobacter hormaechei. We confirmed SCV emergence was attributed to limited genomic mutations: 4 single nucleotide variants in the E. coli SCV, 5 in C. freundii, and 8 in E. hormaechei. In addition, a 10.2 kb chromosomal segment containing 11 genes was deleted in the E. hormaechei SCV isolate. Each SCV had at least one coding change in a gene associated with bacterial oxidative respiration and another involved in iron capture. Chemical and genetic rescue confirmed defects in heme biosynthesis for E. coli and C. freundii and lipoic acid biosynthesis in E. hormaachei were responsible for the SCV phenotype. Prototrophic growth in all 3 SCV Enterobacteriaceae species was unaffected under anaerobic culture conditions in vitro, illustrating how SCVs may persist in vivo.


Asunto(s)
Enterobacteriaceae/genética , Enterobacteriaceae/aislamiento & purificación , Silenciador del Gen , Genes Bacterianos , Hierro/metabolismo , Aerobiosis/genética , Anaerobiosis/genética , Vías Biosintéticas/genética , Niño , Recuento de Colonia Microbiana , Farmacorresistencia Bacteriana/genética , Enterobacteriaceae/crecimiento & desarrollo , Femenino , Variación Genética , Hemo/biosíntesis , Humanos , Lactante , Cinética , Masculino , Pruebas de Sensibilidad Microbiana , Fenotipo , Ácido Tióctico/biosíntesis , Secuenciación Completa del Genoma
16.
Artículo en Inglés | MEDLINE | ID: mdl-33753220

RESUMEN

Molluscs exhibit diverse shell colors. The molecular regulation of shell coloration is however not well understood. To investigate the connection of shell coloration with pigment synthesis, we analyzed the distribution of porphyrins, a widespread group of pigments in nature, in four Pacific oyster strains of different shell colors including black, orange, golden, and white. The porphyrin distribution was analyzed in oyster mantles and shells by fluorescence imaging and UV spectrophotometer. The results showed that red fluorescence emitted by porphyrins under the UV light was detected only on the nacre of the orange-shell strain and mantles of orange, black and white-shell strains. Extracts from newly deposit shell, nacre and mantle tissue from orange-shell specimens showed peaks in UV-vis spectra that are characteristic of porphyrins, but these were not observed for the other shell-color strains. In addition, genes of the haem synthetic pathway were isolated and characterized. Phylogenetic analysis of CgALAS, CgALAD, CgPBGD, CgUROS, and CgUROD provide further evidence for a conserved genetic pathway of haem synthesis during evolution. Differential expression of the haem genes expressed in mantle tissues support these findings and are consistent with porphyrins being produced by the orange strain only. Tissue in situ hybridization demonstrated the expression of these candidate genes at the outer fold of C. gigas mantles where shell is deposited. Our studies provide a better understanding of shell pigmentation in C. gigas and candidate genes for future mechanistic analysis of shell color formation in molluscs.


Asunto(s)
Crassostrea , Hemo , Filogenia , Pigmentación , Animales , Crassostrea/genética , Crassostrea/metabolismo , Hemo/biosíntesis , Hemo/genética
17.
Clin Transl Sci ; 14(4): 1403-1411, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33650309

RESUMEN

Plasma coproporphyrin-I (CP-I) concentration is used as a sensitive and selective endogenous probe for phenotyping organic anion transporting polypeptides 1B (OATP1B) activity in many studies. CP-I is produced in the process of heme synthesis, but the relationship between plasma CP-I concentrations and heme synthesis activity is unknown. In this study, we evaluated the relationship between plasma CP-I concentration and hemoglobin level as a biomarker of heme synthesis activity. The data of 391 subjects selected from the Japanese general population were analyzed. One hundred twenty-six participants had OATP1B1*15 allele, 11 of whom were homozygous (OATP1B1*15/*15). Multiple regression analysis identified hemoglobin level as an independent variable associated with plasma CP-I concentration (p < 0.0001). A significant positive correlation was observed between hemoglobin level and plasma CP-I concentration in participants without OATP1B1*15 allele (n = 265; rs  = 0.35, p < 0.0001) and with OATP1B1*15 allele (n = 126; rs  =0.27, p = 0.0022). However, Kruskal-Wallis test showed no large difference in Kruskal-Wallis statistics between the distribution of plasma CP-I concentrations and that of ratio of plasma CP-I to hemoglobin among six OATP1B1 polymorphism groups. These findings suggest that the hemoglobin level seems to reflect biosynthesis of CP-I. However, correction by hemoglobin level is not required when using basal plasma CP-I concentration for phenotyping OATP1B activity.


Asunto(s)
Coproporfirinas/sangre , Hemoglobinas/análisis , Transportador 1 de Anión Orgánico Específico del Hígado/genética , Adulto , Anciano , Alelos , Biomarcadores/sangre , Estudios de Cohortes , Coproporfirinas/metabolismo , Femenino , Estudio de Asociación del Genoma Completo , Hemo/análisis , Hemo/biosíntesis , Humanos , Transportador 1 de Anión Orgánico Específico del Hígado/metabolismo , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Medicina de Precisión/métodos
18.
Cell Rep ; 34(11): 108869, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33730581

RESUMEN

Mitochondrial carriers (MCs) mediate the passage of small molecules across the inner mitochondrial membrane (IMM), enabling regulated crosstalk between compartmentalized reactions. Despite MCs representing the largest family of solute carriers in mammals, most have not been subjected to a comprehensive investigation, limiting our understanding of their metabolic contributions. Here, we functionally characterize SFXN1, a member of the non-canonical, sideroflexin family. We find that SFXN1, an integral IMM protein with an uneven number of transmembrane domains, is a TIM22 complex substrate. SFXN1 deficiency leads to mitochondrial respiratory chain impairments, most detrimental to complex III (CIII) biogenesis, activity, and assembly, compromising coenzyme Q levels. The CIII dysfunction is independent of one-carbon metabolism, the known primary role for SFXN1 as a mitochondrial serine transporter. Instead, SFXN1 supports CIII function by participating in heme and α-ketoglutarate metabolism. Our findings highlight the multiple ways that SFXN1-based amino acid transport impacts mitochondrial and cellular metabolic efficiency.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Mitocondrias/metabolismo , Transportador 1 de Sodio-Glucosa/metabolismo , Formiatos/farmacología , Eliminación de Gen , Células HEK293 , Células HeLa , Hemo/biosíntesis , Hemina/farmacología , Homeostasis/efectos de los fármacos , Humanos , Hierro/metabolismo , Ácidos Cetoglutáricos/farmacología , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Proteínas del Complejo de Importación de Proteínas Precursoras Mitocondriales/metabolismo , Especificidad por Sustrato/efectos de los fármacos
19.
Mol Biol Evol ; 38(6): 2351-2365, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-33528571

RESUMEN

Blood clams differ from their molluscan kins by exhibiting a unique red-blood (RB) phenotype; however, the genetic basis and biochemical machinery subserving this evolutionary innovation remain unclear. As a fundamental step toward resolving this mystery, we presented the first chromosome-level genome and comprehensive transcriptomes of the blood clam Tegillarca granosa for an integrated genomic, evolutionary, and functional analyses of clam RB phenotype. We identified blood clam-specific and expanded gene families, as well as gene pathways that are of RB relevant. Clam-specific RB-related hemoglobins (Hbs) showed close phylogenetic relationships with myoglobins (Mbs) of blood clam and other molluscs without the RB phenotype, indicating that clam-specific Hbs were likely evolutionarily derived from the Mb lineage. Strikingly, similar to vertebrate Hbs, blood clam Hbs were present in a form of gene cluster. Despite the convergent evolution of Hb clusters in blood clam and vertebrates, their Hb clusters may have originated from a single ancestral Mb-like gene as evidenced by gene phylogeny and synteny analysis. A full suite of enzyme-encoding genes for heme synthesis was identified in blood clam, with prominent expression in hemolymph and resembling those in vertebrates, suggesting a convergence of both RB-related Hb and heme functions in vertebrates and blood clam. RNA interference experiments confirmed the functional roles of Hbs and key enzyme of heme synthesis in the maintenance of clam RB phenotype. The high-quality genome assembly and comprehensive transcriptomes presented herein serve new genomic resources for the super-diverse phylum Mollusca, and provide deep insights into the origin and evolution of invertebrate RB.


Asunto(s)
Arcidae/genética , Evolución Biológica , Hemoglobinas/genética , Animales , Arcidae/metabolismo , Cromosomas , Genoma , Hemo/biosíntesis , Hemolinfa/metabolismo , Humanos , Familia de Multigenes , Transcriptoma
20.
ACS Synth Biol ; 10(2): 402-411, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33497199

RESUMEN

Prokaryotic cell-free coupled transcription-translation (TX-TL) systems are emerging as a powerful tool to examine natural product biosynthetic pathways in a test tube. The key advantages of this approach are the reduced experimental time scales and controlled reaction conditions. To realize this potential, it is essential to develop specialized cell-free systems in organisms enriched for biosynthetic gene clusters. This requires strong protein production and well-characterized synthetic biology tools. The Streptomyces genus is a major source of natural products. To study enzymes and pathways from Streptomyces, we originally developed a homologous Streptomyces cell-free system to provide a native protein folding environment, a high G+C (%) tRNA pool, and an active background metabolism. However, our initial yields were low (36 µg/mL) and showed a high level of batch-to-batch variation. Here, we present an updated high-yield and robust Streptomyces TX-TL protocol, reaching up to yields of 266 µg/mL of expressed recombinant protein. To complement this, we rapidly characterize a range of DNA parts with different reporters, express high G+C (%) biosynthetic genes, and demonstrate an initial proof of concept for combined transcription, translation, and biosynthesis of Streptomyces metabolic pathways in a single "one-pot" reaction.


Asunto(s)
Ingeniería Metabólica/métodos , Familia de Multigenes , Biosíntesis de Proteínas/genética , Streptomyces/genética , Streptomyces/metabolismo , Productos Biológicos/metabolismo , Extractos Celulares , ADN/metabolismo , Hemo/biosíntesis , Melaninas/biosíntesis , Regiones Promotoras Genéticas , Proteínas Recombinantes/metabolismo , Biología Sintética/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA