Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Biochim Biophys Acta Gen Subj ; 1868(11): 130707, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39209088

RESUMEN

Heme is an essential prosthetic molecule for life activities and is well known to act as the active center of many proteins, however, labile heme (LH) released from proteins is a harmful molecule that produces reactive oxygen species and must be strictly controlled. Recently, LH has been suggested to function as an important molecule for diverse physiological responses. Quantitative analysis of the intracellular dynamics of LH is essential for understanding its physiological functions, a substantially practical method has not been established. Here, we successfully developed an alternative method that can be used to complement quantification of the dynamics of intracellular LH using H-FluNox, an activity-based specific fluorescent probe recently constructed. Our newly established method should be effective in elucidating the physiological functions of LH.

2.
Appl Microbiol Biotechnol ; 108(1): 37, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38183476

RESUMEN

A comprehensive analysis to survey heme-binding proteins produced by the white-rot fungus Phanerochaete chrysosporium was achieved using a biotinylated heme-streptavidin beads system. Mitochondrial citrate synthase (PcCS), glyceraldehyde 3-phosphate dehydrogenase (PcGAPDH), and 2-Cys thioredoxin peroxidase (mammalian HBP23 homolog) were identified as putative heme-binding proteins. Among these, PcCS and PcGAPDH were further characterized using heterologously expressed recombinant proteins. Difference spectra of PcCS titrated with hemin exhibited an increase in the Soret absorbance at 414 nm, suggesting that the axial ligand of the heme is a His residue. The activity of PcCS was strongly inhibited by hemin with Ki oxaloacetate of 8.7 µM and Ki acetyl-CoA of 5.8 µM. Since the final step of heme biosynthesis occurred at the mitochondrial inner membrane, the inhibition of PcCS by heme is thought to be a physiological event. The inhibitory mode of the heme was similar to that of CoA analogues, suggesting that heme binds to PcCS at His347 at the AcCoA-CoA binding site, which was supported by the homology model of PcCS. PcGAPDH was also inhibited by heme, with a lower concentration than that for PcCS. This might be caused by the different location of these enzymes. From the integration of these phenomena, it was concluded that metabolic regulations by heme in the central metabolic and heme synthetic pathways occurred in the mitochondria and cytosol. This novel pathway crosstalk between the central metabolic and heme biosynthetic pathways, via a heme molecule, is important in regulating the metabolic balance (heme synthesis, ATP synthesis, flux balance of the tricarboxylic acid (TCA) cycle and cellular redox balance (NADPH production) during fungal aromatic degradation. KEY POINTS: • A comprehensive survey of heme-binding proteins in P. chrysosporium was achieved. • Several heme-binding proteins including CS and GAPDH were identified. • A novel metabolic regulation by heme in the central metabolic pathways was found.


Asunto(s)
Vías Biosintéticas , Phanerochaete , Animales , Hemo , Phanerochaete/genética , Hemina , Proteínas de Unión al Hemo , Mamíferos
3.
Bioinformation ; 19(1): 105-110, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37720292

RESUMEN

Porphyromonas gingivalis, a peripathogen, has several methods to impede or modify the protective mechanisms of the teeth. Targeting the inhibition of the heme protein will prevent the organism from multiplying and inhibit the virulence mechanism. The literature derived oxazole compounds (1-5) were docked against the protein's active site, and the results show that the selected oxazole derivatives exhibit better interaction compared to clinically proven drugs.

4.
mBio ; 14(4): e0132023, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37462360

RESUMEN

Heme, an essential molecule for virtually all living organisms, acts primarily as a cofactor in a large number of proteins. However, how heme is mobilized from the site of synthesis to the locations where hemoproteins are assembled remains largely unknown in cells, especially bacterial ones. In this study, with Shewanella oneidensis as the model, we identified HtpA (SO0126) as a heme-trafficking protein and homolog of TANGO2 proteins found in eukaryotes. We showed that HtpA homologs are widely distributed in all domains of living organisms and have undergone parallel evolution. In its absence, the cytochrome (cyt) c content and catalase activity decreased significantly. We further showed that both HtpA and representative TANGO2 proteins bind heme with 1:1 stoichiometry and a relatively low dissociation constant. Protein interaction analyses substantiated that HtpA directly interacts with the cytochrome c maturation system. Our findings shed light on cross-membrane transport of heme in bacteria and extend the understanding of TANGO2 proteins. IMPORTANCE The intracellular trafficking of heme, an essential cofactor for hemoproteins, remains underexplored even in eukaryotes, let alone bacteria. Here we developed a high-throughput method by which HtpA, a homolog of eukaryotic TANGO2 proteins, was identified to be a heme-binding protein that enhances cytochrome c biosynthesis and catalase activity in Shewanella oneidensis. HtpA interacts with the cytochrome c biosynthesis system directly, supporting that this protein, like TANGO2, functions in intracellular heme trafficking. HtpA homologs are widely distributed, but a large majority of them were found to be non-exchangeable, likely a result of parallel evolution. By substantiating the heme-trafficking nature of HtpA and its eukaryotic homologs, our findings provide general insight into the heme-trafficking process and highlight the functional conservation along evolution in all living organisms.


Asunto(s)
Hemoproteínas , Shewanella , Citocromos c/metabolismo , Hemo/metabolismo , Catalasa/metabolismo , Shewanella/genética , Shewanella/metabolismo , Hemoproteínas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
5.
Biomolecules ; 13(7)2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37509066

RESUMEN

Heme is a double-edged sword. On the one hand, it has a pivotal role as a prosthetic group of hemoproteins in many biological processes ranging from oxygen transport and storage to miRNA processing. On the other hand, heme can transiently associate with proteins, thereby regulating biochemical pathways. During hemolysis, excess heme, which is released into the plasma, can bind to proteins and regulate their activity and function. The role of heme in these processes is under-investigated, with one problem being the lack of knowledge concerning recognition mechanisms for the initial association of heme with the target protein and the formation of the resulting complex. A specific heme-binding sequence motif is a prerequisite for such complex formation. Although numerous short signature sequences indicating a particular protein function are known, a comprehensive analysis of the heme-binding motifs (HBMs) which have been identified in proteins, concerning specific patterns and structural peculiarities, is missing. In this report, we focus on the evaluation of known mammalian heme-regulated proteins concerning specific recognition and structural patterns in their HBMs. The Cys-Pro dipeptide motifs are particularly emphasized because of their more frequent occurrence. This analysis presents a comparative insight into the sequence and structural anomalies observed during transient heme binding, and consequently, in the regulation of the relevant protein.


Asunto(s)
Hemoproteínas , Animales , Proteínas de Unión al Hemo/metabolismo , Fenómenos Biofísicos , Hemoproteínas/genética , Hemoproteínas/metabolismo , Hemo/metabolismo , Unión Proteica , Mamíferos/metabolismo
6.
Int J Biol Sci ; 19(9): 2663-2677, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37324943

RESUMEN

As a peripheral nerve injury disease, cavernous nerve injury (CNI) caused by prostate cancer surgery and other pelvic surgery causes organic damage to cavernous blood vessels and nerves, thereby significantly attenuating the response to phosphodiesterase-5 inhibitors. Here, we investigated the role of heme-binding protein 1 (Hebp1) in erectile function using a mouse model of bilateral CNI, which is known to promote angiogenesis and improve erection in diabetic mice. We found a potent neurovascular regenerative effect of Hebp1 in CNI mice, demonstrating that exogenously delivered Hebp1 improved erectile function by promoting the survival of cavernous endothelial-mural cells and neurons. We further found that endogenous Hebp1 delivered by mouse cavernous pericyte (MCP)-derived extracellular vesicles promoted neurovascular regeneration in CNI mice. Moreover, Hebp1 achieved these effects by reducing vascular permeability through regulation of claudin family proteins. Our findings provide new insights into Hebp1 as a neurovascular regeneration factor and demonstrate its potential therapeutic application to various peripheral nerve injuries.


Asunto(s)
Diabetes Mellitus Experimental , Disfunción Eréctil , Vesículas Extracelulares , Traumatismos de los Nervios Periféricos , Animales , Humanos , Masculino , Diabetes Mellitus Experimental/complicaciones , Modelos Animales de Enfermedad , Disfunción Eréctil/tratamiento farmacológico , Disfunción Eréctil/etiología , Vesículas Extracelulares/metabolismo , Proteínas de Unión al Hemo/farmacología , Regeneración Nerviosa , Pene/irrigación sanguínea , Pene/inervación , Pene/cirugía , Pericitos/metabolismo , Traumatismos de los Nervios Periféricos/terapia
7.
FASEB J ; 37(7): e22981, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37246607

RESUMEN

Oral and gut microbiomes are important for the maintenance of homeostasis in the human body. Altered or disturbed mutualism between their members results in dysbiosis with local injury and subsequent systemic diseases. The high bacterial density causes intense competition among microbiome residents to acquire nutrients, including iron and heme, the latter of high importance for heme auxotrophic members of the Bacteroidetes phylum. Our main hypothesis is that the heme acquisition mechanism, with the leading role played by a novel HmuY family of hemophore-like proteins, can be used to fulfill nutritional requirements and increase virulence. We characterized HmuY homologs expressed by Bacteroides fragilis and compared their properties with the first representative of this family, the HmuY protein of Porphyromonas gingivalis. In contrast to other Bacteroidetes members, B. fragilis produces three HmuY homologs (Bfr proteins). All bfr transcripts were produced at higher levels in bacteria starved of iron and heme (fold change increase ~60, ~90, and ~70 for bfrA, bfrB, and bfrC, respectively). X-ray protein crystallography showed that B. fragilis Bfr proteins are structurally similar to P. gingivalis HmuY and to other homologs, except for differences in the potential heme-binding pockets. BfrA binds heme, mesoheme, and deuteroheme, but preferentially under reducing conditions, using Met175 and Met146 to coordinate heme iron. BfrB binds iron-free protoporphyrin IX and coproporphyrin III, whereas BfrC does not bind porphyrins. HmuY is capable of heme sequestration from BfrA, which might increase the ability of P. gingivalis to cause dysbiosis also in the gut microbiome.


Asunto(s)
Microbioma Gastrointestinal , Porphyromonas gingivalis , Humanos , Bacteroides fragilis/genética , Bacteroides fragilis/metabolismo , Disbiosis , Hemo/metabolismo , Proteínas Bacterianas/metabolismo
8.
Front Cell Infect Microbiol ; 12: 1029833, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36325469

RESUMEN

Flavobacterium columnare, which causes columnaris disease, is one of the costliest pathogens in the freshwater fish-farming industry. The virulence mechanisms of F. columnare are not well understood and current methods to control columnaris outbreaks are inadequate. Iron is an essential nutrient needed for metabolic processes and is often required for bacterial virulence. F. columnare produces siderophores that bind ferric iron for transport into the cell. The genes needed for siderophore production have been identified, but other components involved in F. columnare iron uptake have not been studied in detail. We identified the genes encoding the predicted secreted heme-binding protein HmuY, the outer membrane iron receptors FhuA, FhuE, and FecA, and components of an ATP binding cassette (ABC) transporter predicted to transport ferric iron across the cytoplasmic membrane. Deletion mutants were constructed and examined for growth defects under iron-limited conditions and for virulence against zebrafish and rainbow trout. Mutants with deletions in genes encoding outer membrane receptors, and ABC transporter components exhibited growth defects under iron-limited conditions. Mutants lacking multiple outer membrane receptors, the ABC transporter, or HmuY retained virulence against zebrafish and rainbow trout mirroring that exhibited by the wild type. Some mutants predicted to be deficient in multiple steps of iron uptake exhibited decreased virulence. Survivors of exposure to such mutants were partially protected against later infection by wild-type F. columnare.


Asunto(s)
Enfermedades de los Peces , Infecciones por Flavobacteriaceae , Oncorhynchus mykiss , Animales , Virulencia/genética , Infecciones por Flavobacteriaceae/microbiología , Pez Cebra , Enfermedades de los Peces/microbiología , Flavobacterium/genética , Oncorhynchus mykiss/metabolismo , Oncorhynchus mykiss/microbiología , Sideróforos/genética , Sideróforos/metabolismo , Hierro/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo
9.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 53(6): 1104-1109, 2022 Nov.
Artículo en Chino | MEDLINE | ID: mdl-36443060

RESUMEN

Periodontitis, one of the most common inflammatory oral diseases in human beings, threatens the health of teeth and mouth and is closely associated with the development of many systemic diseases. Existing research about the pathogenesis of periodontitis mainly focuses on the oral microbial homeostasis and its complex interaction with the immune system. Among all the oral microorganisms, Porphyromonas gingivalis ( P. gingivalis) is considered to be the main pathogen causing chronic periodontitis. Recent studies have shown that P. gingivalis poesseses HmuY, a special heme binding protein, which binds with heme to provide essential nutrition for P. gingivalis and activates the host immune system. Therefore, HmuY plays an important role in the growth, proliferation, invasion, and pathogenesis of P. gingivalis and is a potential virulence factor of the bacteria. Existing studies on HmuY are limited to the host immune response that HmuY triggers, and there are still no conclusive findings on whether HmuY participates in the pathogenesis of periodontitis through other ways, such as influencing periodontal bone metabolism. Herein, we reviewed the latest research findings on the biological characteristics and physiological functions of HmuY and its relationship with chronic periodontitis, so as to provide new ideas for in-depth research and further explorations into the pathogenesis of chronic periodontitis.


Asunto(s)
Periodontitis Crónica , Porphyromonas gingivalis , Humanos , Cara , Estado Nutricional
10.
J Fish Dis ; 45(8): 1189-1199, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35671346

RESUMEN

According to the whole-genome bioinformatics analysis, a heme-binding protein from Nocardia seriolae (HBP) was found. HBP was predicted to be a bacterial secretory protein, located at mitochondrial membrane in eukaryotic cells and have a similar protein structure with the heme-binding protein of Mycobacterium tuberculosis, Rv0203. In this study, HBP was found to be a secretory protein and co-localized with mitochondria in FHM cells. Quantitative analysis of mitochondrial membrane potential value, caspase-3 activity, and transcription level of apoptosis-related genes suggested that overexpression of HBP protein can induce cell apoptosis. In conclusion, HBP was a secretory protein which may target to mitochondria and involve in cell apoptosis in host cells. This research will promote the function study of HBP and deepen the comprehension of the virulence factors and pathogenic mechanisms of N. seriolae.


Asunto(s)
Enfermedades de los Peces , Nocardiosis , Nocardia , Animales , Apoptosis , Proteínas Bacterianas/metabolismo , Enfermedades de los Peces/microbiología , Proteínas de Unión al Hemo , Nocardia/genética , Nocardia/metabolismo , Nocardiosis/microbiología , Nocardiosis/veterinaria
11.
Int J Mol Sci ; 22(12)2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-34203861

RESUMEN

Circulating hemopexin is the primary protein responsible for the clearance of heme; therefore, it is a systemic combatant against deleterious inflammation and oxidative stress induced by the presence of free heme. This role of hemopexin is critical in hemolytic pathophysiology. In this review, we outline the current research regarding how the dynamic activity of hemopexin is implicated in sickle cell disease, which is characterized by a pathological aggregation of red blood cells and excessive hemolysis. This pathophysiology leads to symptoms such as acute kidney injury, vaso-occlusion, ischemic stroke, pain crises, and pulmonary hypertension exacerbated by the presence of free heme and hemoglobin. This review includes in vivo studies in mouse, rat, and guinea pig models of sickle cell disease, as well as studies in human samples. In summary, the current research indicates that hemopexin is likely protective against these symptoms and that rectifying depleted hemopexin in patients with sickle cell disease could improve or prevent the symptoms. The data compiled in this review suggest that further preclinical and clinical research should be conducted to uncover pathways of hemopexin in pathological states to evaluate its potential clinical function as both a biomarker and therapy for sickle cell disease and related hemoglobinopathies.


Asunto(s)
Anemia de Células Falciformes/patología , Anemia de Células Falciformes/fisiopatología , Citoprotección , Hemopexina/metabolismo , Animales , Humanos , Inmunomodulación , Lipoproteínas/metabolismo , Microvasos/patología
12.
Int J Biol Macromol ; 184: 955-966, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34153360

RESUMEN

Hemoglobin-based oxygen carriers have long been pursued to meet clinical needs by using native hemoglobin (Hb) from human or animal blood, or recombinantly produced Hb, but the development has been impeded by safety and toxicity issues. Herewith we report the successful production of human fetal hemoglobin (HbF) in Nicotiana benthamiana through Agrobacterium tumefaciens-mediated transient expression. HbF is a heterotetrameric protein composed of two identical α- and two identical γ-subunits, held together by hydrophobic interactions, hydrogen bonds, and salt bridges. In our study, the α- and γ-subunits of HbF were fused in order to stabilize the α-subunits and facilitate balanced expression of α- and γ-subunits in N. benthamiana. Efficient extraction and purification methods enabled production of the recombinantly fused endotoxin-free HbF (rfHbF) in high quantity and quality. The transiently expressed rfHbF protein was identified by SDS-PAGE, Western blot and liquid chromatography-tandem mass spectrometry analyses. The purified rfHbF possessed structural and functional properties similar to native HbF, which were confirmed by biophysical, biochemical, and in vivo animal studies. The results demonstrate a high potential of plant expression systems in producing Hb products for use as blood substitutes.


Asunto(s)
Hemoglobina Fetal/genética , Nicotiana/genética , Oxígeno/metabolismo , Hemoglobina Fetal/aislamiento & purificación , Hemoglobina Fetal/metabolismo , Humanos , Enlace de Hidrógeno , Proteínas de Plantas/aislamiento & purificación , Plantas Modificadas Genéticamente/crecimiento & desarrollo , Plantas Modificadas Genéticamente/metabolismo , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Nicotiana/crecimiento & desarrollo , Nicotiana/metabolismo
13.
Int J Biol Macromol ; 185: 324-337, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34171249

RESUMEN

Mycobacterium tuberculosis, one of the major threats to mankind, requires micronutrients like metal ions for their survival and pathogenicity inside the host system. Intracellular pathogens such as M. tuberculosis have co-evolved to combat the nutritional immunity developed by the host. It has developed eminent mechanisms to sequester essential metal ions from the host system. One such prominent mechanism to scavenge metal ions to thrive in the host cell involves ATP-binding cassette (ABC) transporters, which transport metal ions (in free and/or complex forms) across the cell membrane. This study employs a high-throughput data mining analysis to identify open reading frames (ORFs) encoding metal uptake ABC transporters in M. tuberculosis H37Rv. In total, 19 ORFs resulting in seven ABC transport systems and two P-type ATPases were identified, which are potentially involved in the uptake of different metal ions. The results also suggest the existence of a subunit sharing mechanism in M. tuberculosis where the transmembrane and nucleotide binding domains are shared among different ABC transport systems indicating the import of multiple substrates via a single ABC transporter. Thus, this study reflects an overview of the repertoire of metal-specific ABC transport systems in M. tuberculosis H37Rv, providing potential therapeutic targets for the future.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Metales/metabolismo , Mycobacterium tuberculosis/metabolismo , Transportadoras de Casetes de Unión a ATP/química , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Transporte Biológico , Dominio Catalítico , Minería de Datos , Ligandos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Mycobacterium tuberculosis/genética , Sistemas de Lectura Abierta , Conformación Proteica
14.
Biol Chem ; 402(6): 675-691, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-33581700

RESUMEN

In hemolytic disorders, erythrocyte lysis results in massive release of hemoglobin and, subsequently, toxic heme. Hemopexin is the major protective factor against heme toxicity in human blood and currently considered for therapeutic use. It has been widely accepted that hemopexin binds heme with extraordinarily high affinity of <1 pM in a 1:1 ratio. However, several lines of evidence point to a higher stoichiometry and lower affinity than determined 50 years ago. Here, we re-analyzed these data. SPR and UV/Vis spectroscopy were used to monitor the interaction of heme with the human protein. The heme-binding sites of hemopexin were characterized using hemopexin-derived peptide models and competitive displacement assays. We obtained a KD value of 0.32 ± 0.04 nM and the ratio for the interaction was determined to be 1:1 at low heme concentrations and at least 2:1 (heme:hemopexin) at high concentrations. We were able to identify two yet unknown potential heme-binding sites on hemopexin. Furthermore, molecular modelling with a newly created homology model of human hemopexin suggested a possible recruiting mechanism by which heme could consecutively bind several histidine residues on its way into the binding pocket. Our findings have direct implications for the potential administration of hemopexin in hemolytic disorders.


Asunto(s)
Hemo/química , Hemopexina/química , Humanos , Modelos Moleculares , Espectrofotometría Ultravioleta , Resonancia por Plasmón de Superficie
15.
FEBS Lett ; 595(2): 206-219, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33210733

RESUMEN

Lipocalins are a superfamily of functionally diverse proteins defined by a well-conserved tertiary structure despite variation in sequence. Lipocalins bind and transport small hydrophobic molecules in organisms of all kingdoms. However, there is still uncertainty regarding the function of some members of the family, including bacterial lipocalin Blc from Escherichia coli. Here, we present evidence that lipocalin Blc may be involved in heme binding, trans-periplasmic transport, or heme storage. This conclusion is supported by a cocrystal structure, mass-spectrometric data, absorption titration, and in silico analysis. Binding of heme is observed at low micromolar range with one-to-one ligand-to-protein stoichiometry. However, the absence of classical coordination to the iron atom leaves the possibility that the primary ligand of Blc is another tetrapyrrole.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/química , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Hemo/metabolismo , Lipocalinas/química , Lipocalinas/metabolismo , Cromatografía Liquida , Simulación por Computador , Cristalografía por Rayos X , Hemo/química , Ligandos , Espectrometría de Masas , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Transporte de Proteínas
16.
Pathogens ; 9(4)2020 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-32218184

RESUMEN

Nontypeable Haemophilus influenzae (NTHi) is a leading causative organism of opportunistic respiratory tract infections. However, there are currently no effective vaccination strategies, and existing treatments are compromised by antibiotic resistance. We previously characterized Haemophilus haemolyticus (Hh) strains capable of producing haemophilin (HPL), a heme-binding protein that restricts NTHi growth by limiting its access to an essential growth factor, heme. Thus, these strains may have utility as a probiotic therapy against NTHi infection by limiting colonization, migration and subsequent infection in susceptible individuals. Here, we assess the preliminary feasibility of this approach by direct in vitro competition assays between NTHi and Hh strains with varying capacity to produce HPL. Subsequent changes in NTHi growth rate and fitness, in conjunction with HPL expression analysis, were employed to assess the NTHi-inhibitory capacity of Hh strains. HPL-producing strains of Hh not only outcompeted NTHi during short-term and extended co-culture, but also demonstrated a growth advantage compared with Hh strains unable to produce the protein. Additionally, HPL expression levels during competition correlated with the NTHi-inhibitory phenotype. HPL-producing strains of Hh demonstrate significant probiotic potential against NTHi colonization in the upper respiratory tract, however, further investigations are warranted to demonstrate a range of other characteristics that would support the eventual development of a probiotic.

17.
Curr Genet ; 66(4): 703-711, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32185489

RESUMEN

Iron is essential for nearly all aerobic organisms. One source of iron in nature is in the form of heme. Due to its critical physiological importance as a cofactor for several enzymes, organisms have evolved various means to secure heme for their needs. In the case of heme prototrophs, these organisms possess a highly conserved eight-step biosynthetic pathway. Another means used by many organisms is to acquire heme from external sources. As opposed to the knowledge of enzymes responsible for heme biosynthesis, the nature of the players and mechanisms involved in the acquisition of exogenous heme is limited. This review focuses on a description of newly discovered proteins that have novel functions in heme assimilation in the model organism Schizosaccharomyces pombe. This tractable model allows the use of the power of genetics to selectively block heme biosynthesis, setting conditions to investigate the mechanisms by which external heme is taken up by the cells. Studies have revealed that S. pombe possesses two independent heme uptake systems that require Shu1 and Str3, respectively. Heme-bound iron is captured by Shu1 at the cell surface, triggering its internalization to the vacuole with the aid of ubiquitinated proteins and the ESCRT machinery. In the case of the plasma membrane transporter Str3, it promotes cellular heme import in cells lacking Shu1. The discovery of these two pathways may contribute to gain novel insights into the mechanisms whereby fungi assimilate heme, which is an essentially biological process for their ability to invade and colonize new niches.


Asunto(s)
Hemo/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Animales , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Hongos/metabolismo , Hemo/genética , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo
18.
Front Plant Sci ; 11: 2, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32076429

RESUMEN

Heme plays an active role in primary plant metabolic pathways as well as in stress signaling. In this study, we characterized the predicted heme-binding protein SOUL4. Proteomics evidence suggests that SOUL4 is a component of Arabidopsis plastoglobules (PGs, chloroplast lipid droplets). SOUL4 contains heme-binding motifs and the recombinant protein is shown here to bind heme in vitro. Fluorescence-tagged SOUL4 colocalized with the specific PG marker Fibrillin1A (FBN1A) in transiently transformed Nicotiana benthamiana leaves. In addition, SOUL4 cofractionated with another PG marker Fibrillin2 (FBN2) in sucrose gradient ultracentrifugation experiments. In vitro kinase experiments revealed that SOUL4 is phosphorylated by a yet unknown chloroplast protein kinase. Our data demonstrate that SOUL4 is a bona fide PG protein and may function in heme-buffering in the chloroplast.

19.
Int J Mol Sci ; 19(12)2018 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-30501057

RESUMEN

SOUL, a heme-binding protein-2 (HEBP-2), interacts with apoptosis-linked gene 2 protein (ALG-2) in a Ca2+-dependent manner. To investigate the properties of the interaction of SOUL with ALG-2, we generated several mutants of SOUL and ALG-2 and analyzed the recombinant proteins using pulldown assay and isothermal titration calorimetry. The interaction between SOUL and ALG-2 (delta3-23ALG-2) was an exothermic reaction, with 1:1 stoichiometry and high affinity (Kd = 32.4 nM) in the presence of Ca2+. The heat capacity change (ΔCp) of the reaction showed a large negative value (-390 cal/K·mol), which suggested the burial of a significant nonpolar surface area or disruption of a hydrogen bond network that was induced by the interaction (or both). One-point mutation of SOUL Phe100 or ALG-2 Trp57 resulted in complete loss of heat change, supporting the essential roles of these residues for the interaction. Nevertheless, a truncated mutant of SOUL1-143 that deleted the domain required for the interaction with ALG-2 Trp57 still showed 1:1 binding to ALG-2 with an endothermic reaction. These results provide a better understanding of the target recognition mechanism and conformational change of SOUL in the interaction with ALG-2.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas de Unión al Calcio/metabolismo , Hemoproteínas/metabolismo , Proteínas Gestacionales/metabolismo , Termodinámica , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas de Unión al Calcio/genética , Calorimetría , Cromatografía en Gel , Dicroismo Circular , Proteínas de Unión al Hemo , Hemoproteínas/genética , Ratones , Proteínas Gestacionales/genética , Unión Proteica
20.
Front Microbiol ; 9: 183, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29479345

RESUMEN

The HrrSA and the ChrSA two-component systems play a central role in the coordination of heme homeostasis in the Gram-positive soil bacterium Corynebacterium glutamicum and the prominent pathogen Corynebacterium diphtheriae, both members of the Corynebacteriaceae. In this study, we have performed a comparative analysis of the membrane topology and heme-binding characteristics of the histidine kinases HrrS and ChrS of C. glutamicum. While the cytoplasmic catalytic domains are highly conserved between HrrS and ChrS, the N-terminal sensing parts share only minor sequence similarity. PhoA and LacZ fusions of the N-terminal sensor domains of HrrS and ChrS revealed that both proteins are embedded into the cytoplasmic membrane via six α-helices. Although the overall membrane topology appeared to be conserved, target gene profiling indicated a higher sensitivity of the ChrS system to low heme levels (< 1 µM). In vitro, solubilized and purified full-length proteins bound heme in a 1:1 stoichiometry per monomer. Alanine-scanning of conserved amino acid residues in the N-terminal sensor domain revealed three aromatic residues (Y112, F115, and F118), which apparently contribute to heme binding of HrrS. Exchange of either one or all three residues resulted in an almost abolished heme binding of HrrS in vitro. In contrast, ChrS mutants only displayed a red shift of the soret band from 406 to 418 nm suggesting an altered set of ligands in the triple mutant. In line with target gene profiling, these in vitro studies suggest distinct differences in the heme-protein interface of HrrS and ChrS. Since the membrane topology mapping displayed no extensive loop regions and alanine-scanning revealed potential heme-binding residues in α-helix number four, we propose an intramembrane sensing mechanism for both proteins. Overall, we present a first comparative analysis of the ChrS and HrrS kinases functioning as transient heme sensors in the Corynebacteriaceae.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA