Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(12)2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38928065

RESUMEN

Ferrochelatase (FECH) is the terminal enzyme in human heme biosynthesis, catalyzing the insertion of ferrous iron into protoporphyrin IX (PPIX) to form protoheme IX (Heme). Phosphorylation increases the activity of FECH, and it has been confirmed that the activity of FECH phosphorylated at T116 increases. However, it remains unclear whether the T116 site and other potential phosphorylation modification sites collaboratively regulate the activity of FECH. In this study, we identified a new phosphorylation site, T218, and explored the allosteric effects of unphosphorylated (UP), PT116, PT218, and PT116 + PT218 states on FECH in the presence and absence of substrates (PPIX and Heme) using molecular dynamics (MD) simulations. Binding free energies were evaluated with the MM/PBSA method. Our findings indicate that the PT116 + PT218 state exhibits the lowest binding free energy with PPIX, suggesting the strongest binding affinity. Additionally, this state showed a higher binding free energy with Heme compared to UP, which facilitates Heme release. Moreover, employing multiple analysis methods, including free energy landscape (FEL), principal component analysis (PCA), dynamic cross-correlation matrix (DCCM), and hydrogen bond interaction analysis, we demonstrated that phosphorylation significantly affects the dynamic behavior and binding patterns of substrates to FECH. Insights from this study provide valuable theoretical guidance for treating conditions related to disrupted heme metabolism, such as various porphyrias and iron-related disorders.


Asunto(s)
Dominio Catalítico , Ferroquelatasa , Hemo , Simulación de Dinámica Molecular , Protoporfirinas , Ferroquelatasa/metabolismo , Ferroquelatasa/química , Humanos , Fosforilación , Hemo/metabolismo , Hemo/química , Protoporfirinas/química , Protoporfirinas/metabolismo , Unión Proteica , Sitios de Unión , Termodinámica
2.
Br J Clin Pharmacol ; 89(10): 3092-3104, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37259680

RESUMEN

AIMS: To explore the potential value of serum glutamate dehydrogenase (GLDH), ferrochelatase (FECH), heme oxygenase-1 (HO-1) and glutathione-S-transferase-α (GST-α) as diagnostic biomarkers for liver injury caused by antituberculosis drugs. METHODS: We established a rat model of isoniazide-induced liver injury and recruited 122 hospitalized tuberculosis patients taking antituberculosis drugs. We detected the concentration of GLDH, FECH, HO-1 and GST-α by enzyme-linked immunosorbent assay. GraphPad Prism8 and SPSS 26.0 were used for statistical analysis. RESULTS: In the rat model, serum GLDH concentration gradually increased during isoniazid (INH) administration, while serum FECH, HO-1 and GST-α concentrations significantly increased after INH administration was stopped. The receiver operating characteristic curve showed that the areas under the curve (AUCs) of serum GLDH and FECH for the diagnosis of anti-tuberculosis (TB) drug-induced liver injury (anti-TB-DILI) were 0.7692 (95% confidence interval [CI] 0.5442-0.9943) and 0.7284 (95% CI 0.4863-0.9705) and the diagnostic accuracies were 81.25% and 78.79%, respectively. In clinical research, the AUCs of GLDH and FECH were 0.9124 (95% CI 0.8380-0.9867) and 0.6634 (95% CI 0.5391-0.7877), and the optimal thresholds were 10.40 mIU/mL and 1.304 ng/mL, respectively. The diagnostic accuracy, specificity and positive predictive value (PPV) of GLDH were 82.61%, 79.38% and 47.22%. We performed a joint diagnostic test for GLDH and FECH. The diagnostic accuracy (90.43%), specificity (91.75%) and PPV (65.21%) of serial tests were better than for GLDH and FECH alone. CONCLUSIONS: GLDH in the diagnosis of liver injury induced by anti-TB drugs has high sensitivity, but low specificity and low PPV. The combination of GLDH and FECH could significantly improve the specificity, PPV and diagnostic accuracy, and reduce the false-positive rate of anti-TB-DILI.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Tuberculosis , Ratas , Animales , Antituberculosos/efectos adversos , Glutamato Deshidrogenasa , Ferroquelatasa , Hígado , Biomarcadores , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Tuberculosis/diagnóstico , Tuberculosis/tratamiento farmacológico
3.
Int J Mol Sci ; 24(19)2023 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-37834119

RESUMEN

Sigma receptors are non-opiate/non-phencyclidine receptors that bind progesterone and/or heme and also several unrelated xenobiotics/chemicals. They reside in the plasma membrane and in the membranes of the endoplasmic reticulum, mitochondria, and nucleus. Until recently, the biology/pharmacology of these proteins focused primarily on their role in neuronal functions in the brain/retina. However, there have been recent developments in the field with the discovery of unexpected roles for these proteins in iron/heme homeostasis. Sigma receptor 1 (S1R) regulates the oxidative stress-related transcription factor NRF2 and protects against ferroptosis, an iron-induced cell death process. Sigma receptor 2 (S2R), which is structurally unrelated to S1R, complexes with progesterone receptor membrane components PGRMC1 and PGRMC2. S2R, PGRMC1, and PGRMC2, either independently or as protein-protein complexes, elicit a multitude of effects with a profound influence on iron/heme homeostasis. This includes the regulation of the secretion of the iron-regulatory hormone hepcidin, the modulation of the activity of mitochondrial ferrochelatase, which catalyzes iron incorporation into protoporphyrin IX to form heme, chaperoning heme to specific hemoproteins thereby influencing their biological activity and stability, and protection against ferroptosis. Consequently, S1R, S2R, PGRMC1, and PGRMC2 potentiate disease progression in hemochromatosis and cancer. These new discoveries usher this intriguing group of non-traditional progesterone receptors into an unchartered territory in biology and medicine.


Asunto(s)
Ferroptosis , Receptores sigma , Receptores sigma/metabolismo , Hemo/metabolismo , Receptores de Progesterona/metabolismo , Hierro , Homeostasis
4.
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
5.
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
6.
J Clin Pharm Ther ; 47(8): 1276-1283, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35470464

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: The pathogenic mechanism of anti-tuberculosis drug-induced liver injury (AT-DILI) is still largely unknown. Recent studies have indicated that rifampicin and isoniazid cotreatment causes the accumulation of endogenous protoporphyrin IX in the liver through the haem biosynthesis pathway. Alanine synthase 1 (ALAS1) and ferrochelatase (FECH) are the rate-limiting enzymes in the production of haem. The present study aimed to investigate the genetic contribution of the ALAS1 and FECH genes to the risk of AT-DILI in an Eastern Chinese Han population. METHODS: A 1:4 matched case-control study was conducted, and eight SNPs in the ALAS1 and FECH genes were detected and assessed. A multivariate conditional logistic regression model was used to estimate the association between genotypes and the risk of AT-DILI by the odds ratios (ORs) with 95% confidence intervals (CIs), with liver disease history, hepatoprotectant use, smoking and drinking history as covariates. RESULTS AND DISCUSSION: Overall, 202 AT-DILI cases and 808 controls were included in this study. The female patients carrying polymorphisms of rs11660001 in FECH had an increased risk of AT-DILI under the dominant and additive models (OR = 1.831, 95% CI: 1.014-3.307, p = 0.045; OR = 1.673, 95% CI: 1.015-2.760, p = 0.044, respectively). The peak aspartate transaminase level was significantly higher in female patients carrying the GA+AA genotype of rs11660001 than in those with the GG genotype during anti-TB treatment (p = 0.032). WHAT IS NEW AND CONCLUSION: Based on this 1:4 individual matched case-control study, SNP rs11660001 in the FECH gene may be associated with susceptibility to AT-DILI in Chinese female anti-TB treatment patients. Further studies in larger varied populations are needed to validate our findings.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Tuberculosis , Antituberculosos/efectos adversos , Estudios de Casos y Controles , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Femenino , Ferroquelatasa , Predisposición Genética a la Enfermedad , Hemo , Humanos , Polimorfismo de Nucleótido Simple , Tuberculosis/inducido químicamente , Tuberculosis/tratamiento farmacológico , Tuberculosis/genética
7.
Int J Mol Sci ; 23(13)2022 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-35806474

RESUMEN

During erythropoiesis, there is an enormous demand for the synthesis of the essential cofactor of hemoglobin, heme. Heme is synthesized de novo via an eight enzyme-catalyzed pathway within each developing erythroid cell. A large body of data exists to explain the transcriptional regulation of the heme biosynthesis enzymes, but until recently much less was known about alternate forms of regulation that would allow the massive production of heme without depleting cellular metabolites. Herein, we review new studies focused on the regulation of heme synthesis via carbon flux for porphyrin synthesis to post-translations modifications (PTMs) that regulate individual enzymes. These PTMs include cofactor regulation, phosphorylation, succinylation, and glutathionylation. Additionally discussed is the role of the immunometabolite itaconate and its connection to heme synthesis and the anemia of chronic disease. These recent studies provide new avenues to regulate heme synthesis for the treatment of diseases including anemias and porphyrias.


Asunto(s)
Hemo , Porfirias , Eritropoyesis/fisiología , Regulación de la Expresión Génica , Hemo/metabolismo , Humanos , Porfirias/genética
8.
Kidney Int ; 100(6): 1214-1226, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34534550

RESUMEN

A multitude of disease and therapy related factors drive the frequent development of kidney disorders in cancer patients. Along with chemotherapy, the newer targeted therapeutics can also cause kidney dysfunction through on and off-target mechanisms. Interestingly, among the small molecule inhibitors approved for the treatment of cancers that harbor BRAF-kinase activating mutations, vemurafenib can trigger tubular damage and acute kidney injury. BRAF is a proto-oncogene involved in cell growth. To investigate the underlying mechanisms, we developed cell culture and mouse models of vemurafenib kidney toxicity. At clinically relevant concentrations vemurafenib induces cell-death in transformed and primary mouse and human kidney tubular epithelial cells. In mice, two weeks of daily vemurafenib treatment causes moderate acute kidney injury with histopathological characteristics of kidney tubular epithelial cells injury. Importantly, kidney tubular epithelial cell-specific BRAF gene deletion did not influence kidney function under normal conditions or alter the severity of vemurafenib-associated kidney impairment. Instead, we found that inhibition of ferrochelatase, an enzyme involved in heme biosynthesis contributes to vemurafenib kidney toxicity. Ferrochelatase overexpression protected kidney tubular epithelial cells and conversely ferrochelatase knockdown increased the sensitivity to vemurafenib-induced kidney toxicity. Thus, our studies suggest that vemurafenib-associated kidney tubular epithelial cell dysfunction and kidney toxicity is BRAF-independent and caused, in part, by off-target ferrochelatase inhibition.


Asunto(s)
Ferroquelatasa , Proteínas Proto-Oncogénicas B-raf , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Indoles/toxicidad , Riñón/metabolismo , Ratones , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Sulfonamidas/toxicidad , Vemurafenib
9.
Genet Med ; 23(1): 140-148, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32873934

RESUMEN

PURPOSE: Erythropoietic protoporphyria (EPP), characterized by painful cutaneous photosensitivity, results from pathogenic variants in ferrochelatase (FECH). For 96% of patients, EPP results from coinheriting a rare pathogenic variant in trans of a common hypomorphic variant c.315-48T>C (minor allele frequency 0.05). The estimated prevalence of EPP derived from the number of diagnosed individuals in Europe is 0.00092%, but this may be conservative due to underdiagnosis. No study has estimated EPP prevalence using large genetic data sets. METHODS: Disease-associated FECH variants were identified in the UK Biobank, a data set of 500,953 individuals including 49,960 exome sequences. EPP prevalence was then estimated. The association of FECH variants with EPP-related traits was assessed. RESULTS: Analysis of pathogenic FECH variants in the UK Biobank provides evidence that EPP prevalence is 0.0059% (95% confidence interval [CI]: 0.0042-0.0076%), 1.7-3.0 times more common than previously thought in the UK. In homozygotes for the common c.315-48T>C FECH variant, there was a novel decrement in both erythrocyte mean corpuscular volume (MCV) and hemoglobin. CONCLUSION: The prevalence of EPP has been underestimated secondary to underdiagnosis. The common c.315-48T>C allele is associated with both MCV and hemoglobin, an association that could be important both for those with and without EPP.


Asunto(s)
Protoporfiria Eritropoyética , Bancos de Muestras Biológicas , Europa (Continente) , Ferroquelatasa/genética , Humanos , Mutación , Protoporfiria Eritropoyética/diagnóstico , Protoporfiria Eritropoyética/epidemiología , Protoporfiria Eritropoyética/genética , Reino Unido/epidemiología
10.
BMC Cancer ; 21(1): 971, 2021 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-34461853

RESUMEN

BACKGROUND: Photodynamic therapy with a photosensitizer such as protoporphyrin-IX, a light sensitive metabolite of heme synthesis, is a highly selective treatment for various carcinomas. In previous studies, we found a significant down regulation of the relevant enzyme ferrochelatase in gastrointestinal carcinomas leading to an accumulation of protoporphyrin-IX within the tumor cells. Recent studies showed that a novel anti-cancer drug, Alectinib, an orally available, highly selective, potent second-generation inhibitor of anaplastic lymphoma tyrosinkinase binds to ferrochelatase. Therefore, we were interested to see whether Alectinib treatment might lead to an accumulation of protoporphyrin IX. METHODS: Tumor cells of different origin were cultured, treated with LED-light and Alectinib. Results were gained by flow cytometry, immunohistochemistry and western blotting. Apoptosis was determined by flow cytometric analysis of Annexin V-FITC stained cells. In addition, cells were counterstained with propidium iodide to distinguish early apoptotic cells and late apoptotic/necrotic cells. RESULTS: Here, we report that photodynamic treatment of tumor cell lines of different origin in combination with Alectinib increased protoporphyrin-IX specific fluorescence and concomitantly cell death. CONCLUSIONS: The usage of Alectinib could be another step for enhancing the effectiveness of photodynamic therapy. Further experiments will show whether photodynamic therapy in combination with Alectinib could be a new strategy for the treatment of e.g. peritoneal disseminated carcinomas.


Asunto(s)
Ácido Aminolevulínico/farmacología , Carbazoles/farmacología , Luz , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/farmacología , Piperidinas/farmacología , Protoporfirinas/metabolismo , Fluorescencia , Humanos , Neoplasias/patología , Células Tumorales Cultivadas
11.
J Biol Chem ; 294(29): 11131-11143, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31167780

RESUMEN

Ferrochelatase (FeCh) is an essential enzyme catalyzing the synthesis of heme. Interestingly, in cyanobacteria, algae, and plants, FeCh possesses a conserved transmembrane chlorophyll a/b binding (CAB) domain that resembles the first and the third helix of light-harvesting complexes, including a chlorophyll-binding motif. Whether the FeCh CAB domain also binds chlorophyll is unknown. Here, using biochemical and radiolabeled precursor experiments, we found that partially inhibited activity of FeCh in the cyanobacterium Synechocystis PCC 6803 leads to overproduction of chlorophyll molecules that accumulate in the thylakoid membrane and, together with carotenoids, bind to FeCh. We observed that pigments bound to purified FeCh are organized in an energy-dissipative conformation and further show that FeCh can exist in vivo as a monomer or a dimer depending on its own activity. However, pigmented FeCh was purified exclusively as a dimer. Separately expressed and purified FeCH CAB domain contained a pigment composition similar to that of full-length FeCh and retained its quenching properties. Phylogenetic analysis suggested that the CAB domain was acquired by a fusion between FeCh and a single-helix, high light-inducible protein early in the evolution of cyanobacteria. Following this fusion, the FeCh CAB domain with a functional chlorophyll-binding motif was retained in all currently known cyanobacterial genomes except for a single lineage of endosymbiotic cyanobacteria. Our findings indicate that FeCh from Synechocystis exists mostly as a pigment-free monomer in cells but can dimerize, in which case its CAB domain creates a functional pigment-binding segment organized in an energy-dissipating configuration.


Asunto(s)
Carotenoides/metabolismo , Clorofila A/metabolismo , Clorofila/metabolismo , Ferroquelatasa/metabolismo , Complejos de Proteína Captadores de Luz/metabolismo , Synechocystis/enzimología , Sitios de Unión , Dimerización , Ferroquelatasa/química , Filogenia , Conformación Proteica
12.
Infect Immun ; 88(8)2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32457103

RESUMEN

The human intestinal anaerobic commensal and opportunistic pathogen Bacteroides fragilis does not synthesize the tetrapyrrole protoporphyrin IX in order to form heme that is required for growth stimulation and survival in vivo Consequently, B. fragilis acquires essential heme from host tissues during extraintestinal infection. The absence of several genes necessary for de novo heme biosynthesis is a common characteristic of many anaerobic bacteria; however, the uroS gene, encoding a uroporphyrinogen III synthase for an early step of heme biosynthesis, is conserved among the heme-requiring Bacteroidales that inhabit the mammalian gastrointestinal tract. In this study, we show that the ability of B. fragilis to utilize heme or protoporphyrin IX for growth was greatly reduced in a ΔuroS mutant. This growth defect appears to be linked to the suppression of reverse chelatase and ferrochelatase activities in the absence of uroS In addition, this ΔuroS suppressive effect was enhanced by the deletion of the yifB gene, which encodes an Mg2+-chelatase protein belonging to the ATPases associated with various cellular activities (AAA+) superfamily of proteins. Furthermore, the ΔuroS mutant and the ΔuroS ΔyifB double mutant had a severe survival defect compared to the parent strain in competitive infection assays using animal models of intra-abdominal infection and intestinal colonization. This shows that the presence of the uroS and yifB genes in B. fragilis seems to be linked to pathophysiological and nutritional competitive fitness for survival in host tissues. Genetic complementation studies and enzyme kinetics assays indicate that B. fragilis UroS is functionally different from canonical bacterial UroS proteins. Taken together, these findings show that heme assimilation and metabolism in the anaerobe B. fragilis have diverged from those of aerobic and facultative anaerobic pathogenic bacteria.


Asunto(s)
Proteínas Bacterianas/genética , Infecciones por Bacteroides/microbiología , Bacteroides fragilis/genética , Bacteroides fragilis/patogenicidad , Ferroquelatasa/genética , Hemo/metabolismo , Uroporfirinógeno III Sintetasa/genética , Animales , Proteínas Bacterianas/inmunología , Infecciones por Bacteroides/inmunología , Infecciones por Bacteroides/metabolismo , Infecciones por Bacteroides/patología , Bacteroides fragilis/inmunología , Unión Competitiva , Transporte Biológico , Ferroquelatasa/inmunología , Eliminación de Gen , Regulación de la Expresión Génica , Prueba de Complementación Genética , Hemo/inmunología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Infecciones Intraabdominales/inmunología , Infecciones Intraabdominales/metabolismo , Infecciones Intraabdominales/microbiología , Infecciones Intraabdominales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Ratas Sprague-Dawley , Uroporfirinógeno III Sintetasa/inmunología , Virulencia
13.
FASEB J ; 33(12): 13367-13385, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31553893

RESUMEN

Heme is an essential molecule synthetized through a broadly conserved 8-step route that has been lost in trypanosomatid parasites. Interestingly, Leishmania reacquired by horizontal gene transfer from γ-proteobacteria the genes coding for the last 3 enzymes of the pathway. Here we show that intracellular amastigotes of Leishmania major can scavenge heme precursors from the host cell to fulfill their heme requirements, demonstrating the functionality of this partial pathway. To dissect its role throughout the L. major life cycle, the significance of L. major ferrochelatase (LmFeCH), the terminal enzyme of the route, was evaluated. LmFeCH expression in a heterologous system demonstrated its activity. Knockout promastigotes lacking lmfech were not able to use the ferrochelatase substrate protoporphyrin IX as a source of heme. In vivo infection of Phlebotomus perniciosus with knockout promastigotes shows that LmFeCH is not required for their development in the sandfly. In contrast, the replication of intracellular amastigotes was hampered in vitro by the deletion of lmfech. However, LmFeCH-/- parasites produced disease in a cutaneous leishmaniasis murine model in a similar way as control parasites. Therefore, although L. major can synthesize de novo heme from macrophage precursors, this activity is dispensable being an unsuited target for leishmaniasis treatment.-Orrego, L. M., Cabello-Donayre, M., Vargas, P., Martínez-García, M., Sánchez, C., Pineda-Molina, E., Jiménez, M., Molina, R., Pérez-Victoria, J. M. Heme synthesis through the life cycle of the heme auxotrophic parasite Leishmania major.


Asunto(s)
Ferroquelatasa/metabolismo , Hemo/biosíntesis , Leishmania major/crecimiento & desarrollo , Leishmaniasis Cutánea/metabolismo , Proteínas Protozoarias/metabolismo , Psychodidae/metabolismo , Virulencia , Secuencia de Aminoácidos , Animales , Coproporfirinógeno Oxidasa/metabolismo , Femenino , Ferroquelatasa/química , Ferroquelatasa/genética , Leishmaniasis Cutánea/parasitología , Macrófagos/metabolismo , Macrófagos/parasitología , Masculino , Ratones , Ratones Endogámicos BALB C , Conformación Proteica , Protoporfirinógeno-Oxidasa/metabolismo , Proteínas Protozoarias/química , Proteínas Protozoarias/genética , Psychodidae/parasitología , Homología de Secuencia
14.
Int J Mol Sci ; 21(20)2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33066693

RESUMEN

Isatin (indole-2, 3-dione) is a non-peptide endogenous bioregulator exhibiting a wide spectrum of biological activity, realized in the cell via interactions with numerous isatin-binding proteins, their complexes, and (sub) interactomes. There is increasing evidence that isatin may be involved in the regulation of complex formations by modulating the affinity of the interacting protein partners. Recently, using Surface Plasmon Resonance (SPR) analysis, we have found that isatin in a concentration dependent manner increased interaction between two human mitochondrial proteins, ferrochelatase (FECH), and adrenodoxine reductase (ADR). In this study, we have investigated the affinity-enhancing effect of isatin on the FECH/ADR interaction. The SPR analysis has shown that FECH forms not only homodimers, but also FECH/ADR heterodimers. The affinity-enhancing effect of isatin on the FECH/ADR interaction was highly specific and was not reproduced by structural analogues of isatin. Bioinformatic analysis performed using three dimensional (3D) models of the interacting proteins and in silico molecular docking revealed the most probable mechanism involving FECH/isatin/ADR ternary complex formation. In this complex, isatin is targeted to the interface of interacting FECH and ADR monomers, forming hydrogen bonds with both FECH and ADR. This is a new regulatory mechanism by which isatin can modulate protein-protein interactions (PPI).


Asunto(s)
Ferredoxina-NADP Reductasa/química , Ferroquelatasa/química , Isatina/química , Ferredoxina-NADP Reductasa/metabolismo , Ferroquelatasa/metabolismo , Humanos , Isatina/metabolismo , Simulación del Acoplamiento Molecular , Unión Proteica , Resonancia por Plasmón de Superficie
15.
Int J Mol Sci ; 21(8)2020 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-32331454

RESUMEN

Exogenous administration of hexaminolevulinate (HAL) induces fluorescent protoporphyrin IX (PpIX) accumulation preferentially in cancer cells. However, the PpIX fluorescence intensities between noncancer and cancer cells are highly variable. The contrast between cancer and noncancer cells may be insufficient to reliably discriminate, especially at the single cell level in cancer diagnostics. This study examines the use of the chemical adjuvants dimethylsulphoxide (DMSO) or deferoxamine (DFO) to enhance the HAL induced PpIX accumulation in cancer cells. Our results showed that in some of the incubation conditions tested, the addition of DFO with HAL significantly increased PpIX 21 fluorescence of adherent monolayer cancer cells, but this was never the case for cells in suspension. Permeabilisation with DMSO did not increase PpIX fluorescence. Cell-to-cell interaction may well play an important role in the PpIX accumulation when suspended cells are treated in HAL and adjuvant chemicals.


Asunto(s)
Ácido Aminolevulínico/análogos & derivados , Fluorescencia , Imagen Molecular , Fármacos Fotosensibilizantes/metabolismo , Protoporfirinas/metabolismo , Ácido Aminolevulínico/metabolismo , Vías Biosintéticas , Línea Celular Tumoral , Hemo/biosíntesis , Humanos
16.
Physiol Mol Biol Plants ; 26(8): 1551-1568, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32801486

RESUMEN

ABSTRACT: Chickpea (Cicer arietinum) belonging to the Fabaceae family is a major legume crop and is a good source of protein and carbohydrates. Industrialization has resulted in soil contamination with heavy metals such as cadmium. Adsorption of cadmium by plants can lead to reduced yields and heavy metal toxicity. In the current study, changes in the anatomical, morphological features and biochemical properties of the chickpea plant were evaluated. Two indexes DWSTI and PHSTI were determined. Anatomically, there was a change in the number of xylem poles within the root structure which was most significant at treatments of 125 µg cadmium. There was also a noticeable change in leaf pigmentation, the total phenolics and soluble protein in the plant. Cadmium levels were elevated attaining concentrations of 0.21, 0.40 and 0.52 mg per gram dry weight in plants exposed to 62, 125 and 250 µg/g Perlit cadmium after a period of 30 days. A noticeable increase in the level of cadmium in the plant was observed. Two PCS genes, glutathione gamma-glutamylcysteinyltransferase 1 and glutathione gamma-glutamylcysteinyltransferase and four FC genes, 4 proteins and 4 mRNA were detected in chickpeas. Bioinformatics tools were utilized to predict enzyme structure and binding sites. Chickpea may be classified as a cadmium hyperaccumulator and may be considered for use in phytoremediation. This study provides a better understanding with regards to the response of chickpeas to cadmium and the genetic mechanism by which the plant regulates heavy metal toxicity.

17.
Mol Genet Metab ; 128(3): 358-362, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30454868

RESUMEN

The erythropoietic porphyrias are inborn errors of heme biosynthesis with prominent cutaneous manifestations. They include autosomal recessive Congenital Erythropoietic Porphyria (CEP) due to loss-of-function (LOF) mutations in the Uroporphyrinogen III Synthase (UROS) gene, Erythropoietic Protoporphyria (EPP) due to LOF mutations in the ferrochelatase (FECH) gene, and X-Linked Protoporphyria (XLP) due to gain-of-function mutations in the terminal exon of the Aminolevulinic Acid Synthase 2 (ALAS2) gene. During the 11-year period from 01/01/2007 through 12/31/2017, the Mount Sinai Porphyrias Diagnostic Laboratory provided molecular diagnostic testing for one or more of these disorders in 628 individuals, including 413 unrelated individuals. Of these 628, 120 patients were tested for CEP, 483 for EPP, and 331 for XLP, for a total of 934 tests. For CEP, 24 of 78 (31%) unrelated individuals tested had UROS mutations, including seven novel mutations. For EPP, 239 of 362 (66%) unrelated individuals tested had pathogenic FECH mutations, including twenty novel mutations. The IVS3-48 T > C low-expression allele was present in 231 (97%) of 239 mutation-positive EPP probands with a pathogenic FECH mutation. In the remaining 3%, three patients with two different FECH mutations in trans were identified. For XLP, 24 of 250 (10%) unrelated individuals tested had ALAS2 exon 11 mutations. No novel ALAS2 mutations were identified. Among family members referred for testing, 33 of 42 (79%) CEP, 62 of 121 (51%) EPP, and 31 of 81 (38%) XLP family members had the respective family mutation. Mutation-positive CEP, EPP, and XLP patients who had been biochemically tested had marked elevations of the disease-appropriate porphyrin intermediates. These results expand the molecular heterogeneity of the erythropoietic porphyrias by adding a total of 27 novel mutations. The results document the usefulness of molecular testing to confirm the positive biochemical findings in these patients and to identify heterozygous family members.


Asunto(s)
Ferroquelatasa/genética , Heterogeneidad Genética , Mutación , Porfiria Eritropoyética/genética , Protoporfiria Eritropoyética/genética , Uroporfirinógeno III Sintetasa/genética , Familia , Femenino , Tamización de Portadores Genéticos , Hemo/biosíntesis , Humanos , Masculino , Técnicas de Diagnóstico Molecular , Trastornos por Fotosensibilidad/etiología , Protoporfiria Eritropoyética/diagnóstico
18.
BMC Microbiol ; 19(1): 173, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31362704

RESUMEN

BACKGROUND: Most species of Shewanella harbor two ferrochelatase paralogues for the biosynthesis of c-type cytochromes, which are crucial for their respiratory versatility. In our previous study of the Shewanella loihica PV-4 strain, we found that the disruption of hemH1 but not hemH2 resulted in a significant accumulation of extracellular protoporphyrin IX (PPIX), but it is different in Shewanella oneidensis MR-1. Hence, the function and transcriptional regulation of two ferrochelatase genes, hemH1 and hemH2, are investigated in S. oneidensis MR-1. RESULT: In the present study, deletion of either hemH1 or hemH2 in S. oneidensis MR-1 did not lead to overproduction of extracellular protoporphyrin IX (PPIX) as previously described in the hemH1 mutants of S. loihica PV-4. Moreover, supplement of exogenous hemins made it possible to generate the hemH1 and hemH2 double mutant in MR-1, but not in PV-4. Under aerobic condition, exogenous hemins were required for the growth of MR-1ΔhemH1ΔhemH2, which also overproduced extracellular PPIX. These results suggest that heme is essential for aerobic growth of Shewanella species and MR-1 could also uptake hemin for biosynthesis of essential cytochrome(s) and respiration. Besides, the exogenous hemin mediated CymA cytochrome maturation and the cellular KatB catalase activity. Both hemH paralogues were transcribed in wild-type MR-1, and the hemH2 transcription was remarkably up-regulated in MR-1ΔhemH1 mutant to compensate for the loss of hemH1. The periplasmic glutathione peroxidase gene pgpD, located in the same operon with hemH2, and a large gene cluster coding for iron, heme (hemin) uptake systems are absent in the PV-4 genome. CONCLUSION: Our results indicate that the genetic divergence in gene content and gene expression between these Shewanella species, accounting for the phenotypic difference described here, might be due to their speciation and adaptation to the specific habitats (iron-rich deep-sea vent versus iron-poor freshwater) in which they evolved and the generated mutants could potentially be utilized for commercial production of PPIX.


Asunto(s)
Citocromos/metabolismo , Ferroquelatasa/genética , Hemo/metabolismo , Protoporfirinas/metabolismo , Shewanella , Proteínas Bacterianas/genética , Ecosistema , Agua Dulce/química , Agua Dulce/microbiología , Expresión Génica , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Genotipo , Glutatión Peroxidasa/genética , Hemoproteínas/metabolismo , Hierro/metabolismo , Fenotipo , Agua de Mar/química , Agua de Mar/microbiología , Shewanella/genética , Shewanella/metabolismo
19.
Plant Cell Environ ; 42(2): 618-632, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30242849

RESUMEN

Ferrochelatase (FC) is the final enzyme for haem formation in the tetrapyrrole biosynthesis pathway and encoded by two genes in higher plants. FC2 exists predominantly in green tissue, whereas FC1 is constitutively expressed. We intended to substantiate the specific roles of FC1. The embryo-lethal fc1-2 mutant was used to express the two genomic FC-encoding sequences under the FC1 and FC2 promoter and explore the complementation of the FC1 deficiency. Apart from the successful complementation with FC1, expression of FC2 under control of the FC1 promoter (pFC1::FC2) compensates for missing FC1 but not by FC2 promoter expression. The complementing lines pFC1FC2(fc1/fc1) succeeded under standard growth condition but failed under salt stress. The pFC1FC2(fc1/fc1) line exhibited symptoms of leaf senescence, including accelerated loss of haem and chlorophyll and elevated gene expression for chlorophyll catabolism. In contrast, ectopic FC1 expression (p35S::FC1) resulted in increased chlorophyll accumulation. The limited ability of FC2 to complement fc1 is explained by a faster turnover of FC2 mRNA during stress. It is suggested that FC1-produced haem is essential for embryogenesis and stress response. The pFC1::FC2 expression readily complements the fc1-2 embryo lethality, whereas higher FC1 transcript content contributes essentially to stress tolerance.


Asunto(s)
Proteínas de Arabidopsis/fisiología , Arabidopsis/enzimología , Ferroquelatasa/fisiología , Semillas/crecimiento & desarrollo , Arabidopsis/genética , Arabidopsis/crecimiento & desarrollo , Arabidopsis/fisiología , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Clorofila/metabolismo , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Hemo/metabolismo , Metiltransferasas/metabolismo , Plantas Modificadas Genéticamente , Reacción en Cadena en Tiempo Real de la Polimerasa , Semillas/enzimología , Tilacoides/metabolismo
20.
Acta Derm Venereol ; 99(7): 634-639, 2019 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-30938825

RESUMEN

Erythropoietic protoporphyria is caused by a partial deficiency of ferrochelatase, which is the last enzyme in the heme biosynthesis pathway. In a typical erythropoietic protoporphyria, photosensitivity initially appears, following the first exposure to the sun in early infancy or childhood. Erythropoietic protoporphyria has been reported worldwide, but there is a regional variation in its epidemiology. Approximately 20% of the Japanese patients were recognized to have symptoms of erythropoietic protoporphyria after 10 years of age. Physicians occasionally encounter Japanese patients with erythropoietic protoporphyria, mild symptoms and no FECH gene mutations. The homozygous IVS3-48C polymorphism may cause a mild phenotype of the erythropoietic protoporphyria via a slight increase in protoporphyrin. The frequency of the homozygous IVS3-48C polymorphism in the Japanese population is higher than that observed in European countries. Japanese type of erythropoietic protopor-phyria shows a characteristic phenotype of the late onset and mild symptoms compared to the Caucasian erythropoietic protoporphyria. This review describes the characteristics of erythropoietic protoporphyria in Japanese patients.


Asunto(s)
Ferroquelatasa/genética , Protoporfiria Eritropoyética/epidemiología , Protoporfiria Eritropoyética/genética , Edad de Inicio , Anemia/etiología , Colelitiasis/etiología , Europa (Continente)/epidemiología , Homocigoto , Humanos , Japón/epidemiología , Hepatopatías/etiología , Mutación , América del Norte/epidemiología , Fenotipo , Polimorfismo Genético , Prevalencia , Protoporfiria Eritropoyética/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA