Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
1.
Nat Commun ; 15(1): 3976, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38729948

RESUMEN

Bleeding and thrombosis are known as common complications of polycythemia for a long time. However, the role of coagulation system in erythropoiesis is unclear. Here, we discover that an anticoagulant protein tissue factor pathway inhibitor (TFPI) plays an essential role in erythropoiesis via the control of heme biosynthesis in central macrophages. TFPI levels are elevated in erythroblasts of human erythroblastic islands with JAK2V617F mutation and hypoxia condition. Erythroid lineage-specific knockout TFPI results in impaired erythropoiesis through decreasing ferrochelatase expression and heme biosynthesis in central macrophages. Mechanistically, the TFPI interacts with thrombomodulin to promote the downstream ERK1/2-GATA1 signaling pathway to induce heme biosynthesis in central macrophages. Furthermore, TFPI blockade impairs human erythropoiesis in vitro, and normalizes the erythroid compartment in mice with polycythemia. These results show that erythroblast-derived TFPI plays an important role in the regulation of erythropoiesis and reveal an interplay between erythroblasts and central macrophages.


Asunto(s)
Eritroblastos , Eritropoyesis , Factor de Transcripción GATA1 , Hemo , Lipoproteínas , Macrófagos , Policitemia , Policitemia/metabolismo , Policitemia/genética , Policitemia/patología , Eritroblastos/metabolismo , Hemo/metabolismo , Humanos , Animales , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Ratones , Factor de Transcripción GATA1/metabolismo , Factor de Transcripción GATA1/genética , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Trombomodulina/metabolismo , Trombomodulina/genética , Ratones Noqueados , Ferroquelatasa/metabolismo , Ferroquelatasa/genética , Masculino , Sistema de Señalización de MAP Quinasas , Ratones Endogámicos C57BL , Femenino
2.
Tohoku J Exp Med ; 261(2): 117-122, 2023 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-37495523

RESUMEN

Erythropoietic protoporphyria (EPP) is a very rare disease with an estimated prevalence of 1 in 200,000 individuals. Decreased ferrochelatase activity causes the accumulation of protoporphyrin in the body, and light exposure results in the generation of active oxygen, causing photosensitivity. Liver damage has the greatest influence on the prognosis, and liver transplantation is the only treatment option for patients with decompensated liver cirrhosis. We report a case of living-donor liver transplantation for decompensated liver cirrhosis associated with EPP. The patient was a 52-year-old male who led a normal life except for mild photosensitivity. When the patient was 37-year-old, hepatic dysfunction was noticed. At 48-year-old, high erythrocyte protoporphyrin levels, skin biopsy, and genetic tests resulted in a diagnosis of EPP. The patient underwent living- donor liver transplantation because of decompensated liver cirrhosis. In the operating room and intensive care unit, a special light-shielding film was applied to all light sources to block light with harmful wavelengths during treatment. Due to the need for special measures, a lecture on patients with EPP was given before surgery to deepen understanding among all medical professionals involved in the treatment. As a result, no adverse events occurred during the perioperative period, and the patient was discharged on the 46th post-operative day. Currently, the transplanted liver is functioning extremely well, and the patient is alive 3 years post-transplant. Herein, we describe a case of living donor liver transplantation for EPP with a brief literature review.


Asunto(s)
Hepatopatías , Trasplante de Hígado , Protoporfiria Eritropoyética , Masculino , Humanos , Persona de Mediana Edad , Adulto , Protoporfiria Eritropoyética/cirugía , Protoporfiria Eritropoyética/complicaciones , Protoporfiria Eritropoyética/genética , Trasplante de Hígado/efectos adversos , Donadores Vivos , Protoporfirinas , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Hepatopatías/complicaciones , Cirrosis Hepática/complicaciones , Cirrosis Hepática/cirugía
3.
Oncol Rep ; 48(4)2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36082808

RESUMEN

Iron is an essential nutrient that facilitates cell proliferation and growth, and it can contribute to tumor growth. Although iron chelators have shown great potential in preclinical cancer models, they can cause adverse side­effects. The aim of the present study was to determine whether treatment with 5­aminolevurinic acid (5­ALA) has antitumor effects in bladder cancer, by reduction of mitochondrial iron without using an iron chelator, through activation of heme synthesis. T24 and MGH­U3 cells were treated with 5­ALA. Ferrochelatase uses iron to convert protoporphyrin IX into heme, thus additional groups of T24 and MGH­U3 cells were transfected with synthesized ferrochelatase small interfering RNA (siRNA) either to silence ferrochelatase or to provide a negative siRNA control group, and then cell viability, apoptosis, mitochondrial Fe2+, the cell cycle, and ferritin expression were analyzed in all groups and compared. As an in vivo assessment, mice with orthotopic bladder cancer induced using N­butyl­N­(4­hydro­oxybutyl) were treated with 5­ALA. Bladder weight and pathological findings were evaluated, and immunohistochemical analysis was performed for ferritin and proliferating cell nuclear antigen (PCNA). In the cells treated with 5­ALA, proliferation was decreased compared with the controls, and apoptosis was not detected. In addition, the expression of Fe2+ in mitochondria was decreased by 5­ALA, expression of ferritin was also reduced by 5­ALA, and the percentage of cells in the S phase of the cell cycle was significantly increased by 5­ALA. In T24 and MGH­U3 cells with silenced ferrochelatase, the inhibition of cell proliferation, decreased expression of Fe2+ in mitochondria, reduced expression of ferritin, and increased percentage of cells in the S phase by treatment with 5­ALA were weakened. In vivo, no mouse treated with 5­ALA developed muscle­invasive bladder cancer. The expression of ferritin was weaker in mice treated with 5­ALA and that of PCNA was higher than that in mice treated without 5­ALA. It was concluded that 5­ALA inhibited proliferation of bladder cancer cells by activating heme synthesis.


Asunto(s)
Ferroquelatasa , Neoplasias de la Vejiga Urinaria , Ácido Aminolevulínico/farmacología , Ácido Aminolevulínico/uso terapéutico , Animales , Proliferación Celular , Ferritinas , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Hemo/metabolismo , Hierro/metabolismo , Ratones , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Interferente Pequeño , Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética
4.
Photodiagnosis Photodyn Ther ; 37: 102629, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34798346

RESUMEN

BACKGROUND: Erythropoietic protoporphyria (EPP) is a rare genetic photodermatosis caused by loss-of-function mutations in the gene for ferrochelatase leading to accumulation of the fluorescent protoporphyrin IX (PpIX) in erythrocytes. The mutations are most often inherited mutations present in all cells causing inherited EPP. In very rare cases EPP are acquired in association with myelodysplastic syndromes or myeloproliferative neoplasms, conditions with genetic instability. CASE REPORT: We report a case of acquired EPP in association with hematological disease. We followed erythrocyte PpIX concentration over a year and measured PpIX fluorescence in individual erythrocytes in a blood sample from the case using flow cytometry. The major proportion of erythrocytes did not fluoresce (84%), whereas 13% contained low PpIX fluorescence, 1% contained medium fluorescence, and 2% contained high fluorescence. DISCUSSION: Our observation of the very skewed PpIX distribution in erythrocytes supports the description that acquired EPP is caused by a somatic mutation effecting a clone of hematopoietic cells.


Asunto(s)
Fotoquimioterapia , Protoporfiria Eritropoyética , Eritrocitos , Ferroquelatasa/genética , Humanos , Fotoquimioterapia/métodos , Protoporfiria Eritropoyética/genética , Protoporfirinas/metabolismo
5.
Sci Rep ; 11(1): 20943, 2021 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-34686726

RESUMEN

Non-functioning pituitary adenomas (NFPAs) are typical pituitary macroadenomas in adults associated with increased mortality and morbidity. Although pituitary adenomas are commonly considered slow-growing benign brain tumors, numerous of them possess an invasive nature. Such tumors destroy sella turcica and invade the adjacent tissues such as the cavernous sinus and sphenoid sinus. In these cases, the most critical obstacle for complete surgical removal is the high risk of damaging adjacent vital structures. Therefore, the development of novel therapeutic strategies for either early diagnosis through biomarkers or medical therapies to reduce the recurrence rate of NFPAs is imperative. Identification of gene interactions has paved the way for decoding complex molecular mechanisms, including disease-related pathways, and identifying the most momentous genes involved in a specific disease. Currently, our knowledge of the invasion of the pituitary adenoma at the molecular level is not sufficient. The current study aimed to identify critical biomarkers and biological pathways associated with invasiveness in the NFPAs using a three-way interaction model for the first time. In the current study, the Liquid association method was applied to capture the statistically significant triplets involved in NFPAs invasiveness. Subsequently, Random Forest analysis was applied to select the most important switch genes. Finally, gene set enrichment (GSE) and gene regulatory network (GRN) analyses were applied to trace the biological relevance of the statistically significant triplets. The results of this study suggest that "mRNA processing" and "spindle organization" biological processes are important in NFAPs invasiveness. Specifically, our results suggest Nkx3-1 and Fech as two switch genes in NFAPs invasiveness that may be potential biomarkers or target genes in this pathology.


Asunto(s)
Adenoma/genética , Ferroquelatasa/genética , Genes de Cambio/genética , Proteínas de Homeodominio/genética , Invasividad Neoplásica/genética , Neoplasias Hipofisarias/genética , Factores de Transcripción/genética , Adenoma/patología , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Humanos , Invasividad Neoplásica/patología , Neoplasias Hipofisarias/patología , ARN Mensajero/genética , Silla Turca/patología
6.
Int J Mol Sci ; 21(9)2020 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-32397263

RESUMEN

Photodynamic therapy (PDT) has been used to treat certain types of non-melanoma skin cancer with promising results. However, some skin lesions have not fully responded to this treatment, suggesting a potential PDT-resistant phenotype. Therefore, novel therapeutic alternatives must be identified that improve PDT in resistant skin cancer. In this study, we analyzed the cell viability, intracellular protoporphyrin IX (PpIX) content and subcellular localization, proliferation profile, cell death, reactive oxygen species (ROS) detection and relative gene expression in PDT-resistant HSC-1 cells. PDT-resistant HSC-1 cells show a low quantity of protoporphyrin IX and low levels of ROS, and thus a low rate of death cell. Furthermore, the resistant phenotype showed a downregulation of HSPB1, SLC15A2, FECH, SOD2 and an upregulation of HMBS and BIRC5 genes. On the other hand, epigallocatechin gallate catechin enhanced the MAL-PDT effect, increasing levels of protoporphyrin IX and ROS, and killing 100% of resistant cells. The resistant MAL-PDT model of skin cancer squamous cells (HSC-1) is a reliable and useful tool to understand PDT cytotoxicity and cellular response. These resistant cells were successfully sensitized with epigallocatechin gallate catechin. The in vitro epigallocatechin gallate catechin effect as an enhancer of MAL-PDT in resistant cells is promising in the treatment of difficult skin cancer lesions.


Asunto(s)
Anticarcinógenos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Catequina/análogos & derivados , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Terapia Combinada/métodos , Fotoquimioterapia/métodos , Neoplasias Cutáneas/tratamiento farmacológico , Ácido Aminolevulínico/análogos & derivados , Ácido Aminolevulínico/farmacología , Carcinoma de Células Escamosas/radioterapia , Catequina/farmacología , Muerte Celular/efectos de la radiación , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Hipoxia de la Célula/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Fármacos Fotosensibilizantes/metabolismo , Protoporfirinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Cutáneas/radioterapia , Estrés Fisiológico/efectos de los fármacos , Estrés Fisiológico/genética , Estrés Fisiológico/efectos de la radiación , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Survivin/genética , Survivin/metabolismo , Simportadores/genética , Simportadores/metabolismo
7.
Nucleic Acids Res ; 48(9): 4658-4671, 2020 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-32313951

RESUMEN

Erythropoietic protoporphyria (EPP) is a rare genetic disease in which patients experience acute phototoxic reactions after sunlight exposure. It is caused by a deficiency in ferrochelatase (FECH) in the heme biosynthesis pathway. Most patients exhibit a loss-of-function mutation in trans to an allele bearing a SNP that favors aberrant splicing of transcripts. One viable strategy for EPP is to deploy splice-switching oligonucleotides (SSOs) to increase FECH synthesis, whereby an increase of a few percent would provide therapeutic benefit. However, successful application of SSOs in bone marrow cells is not described. Here, we show that SSOs comprising methoxyethyl-chemistry increase FECH levels in cells. We conjugated one SSO to three prototypical targeting groups and administered them to a mouse model of EPP in order to study their biodistribution, their metabolic stability and their FECH splice-switching ability. The SSOs exhibited distinct distribution profiles, with increased accumulation in liver, kidney, bone marrow and lung. However, they also underwent substantial metabolism, mainly at their linker groups. An SSO bearing a cholesteryl group increased levels of correctly spliced FECH transcript by 80% in the bone marrow. The results provide a promising approach to treat EPP and other disorders originating from splicing dysregulation in the bone marrow.


Asunto(s)
Ferroquelatasa/genética , Oligonucleótidos/administración & dosificación , Protoporfiria Eritropoyética/metabolismo , Empalme del ARN , Albúminas/metabolismo , Animales , Médula Ósea/metabolismo , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Ferroquelatasa/metabolismo , Humanos , Células K562 , Ratones , Oligonucleótidos/sangre , Oligonucleótidos/química , Oligonucleótidos/farmacocinética , Polimorfismo de Nucleótido Simple , Protoporfiria Eritropoyética/genética , Protoporfiria Eritropoyética/terapia , Sitios de Empalme de ARN , Distribución Tisular
9.
Photodermatol Photoimmunol Photomed ; 36(1): 29-33, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31374130

RESUMEN

BACKGROUND: Erythropoietic protoporphyria (EPP) is a semi-dominantly inherited porphyria presenting with photosensitivity during early childhood. Acquired EPP has been reported; however, data regarding this rare disorder are scarce. PURPOSE: To evaluate the characteristics of acquired EPP. METHODS: A comprehensive search of PubMed, Google Scholar, ScienceDirect, and clinicaltrials.gov databases was performed by three reviewers. Studies describing patients with acquired EPP were included. Additionally, we present an index case of a 26-year-old patient who acquired clinically and biochemically typical EPP in association with myelodysplastic syndrome (MDS). RESULTS: We included 20 case reports describing 20 patients. Most (80%) patients were male of mean age 58 ± 13 years. In all patients, acquired EPP was associated with hematological disease, most commonly MDS (85%) followed by myeloproliferative disease (10%). In 86% of cases, hematological disease led to abnormality or somatic mutation in chromosome 18q (the locus of the ferrochelatase gene). The mean erythrocyte protoporphyrin IX concentration was very high (4286 µg/dL). Most (90%) patients presented with photosensitivity, 20% experienced blistering, and 25% presented with hepatic insufficiency, both uncommon in EPP. In 55% of patients, hematological disease was diagnosed after occurrence of cutaneous symptoms. Beta-carotene led to partial control of symptoms in 5 patients and resolution in another patient. Azacitidine treatment of MDS led to resolution of cutaneous symptoms in three patients. CONCLUSION: We present the distinct features of acquired EPP and highlight that any patient presenting with new-onset photosensitivity, irrespective of age should be evaluated for porphyria.


Asunto(s)
Azacitidina/uso terapéutico , Síndromes Mielodisplásicos , Trastornos por Fotosensibilidad , Protoporfiria Eritropoyética , beta Caroteno/uso terapéutico , Adulto , Anciano , Cromosomas Humanos Par 18/genética , Cromosomas Humanos Par 18/metabolismo , Eritrocitos/metabolismo , Femenino , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Sitios Genéticos , Humanos , Masculino , Persona de Mediana Edad , Mutación , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Trastornos por Fotosensibilidad/inducido químicamente , Trastornos por Fotosensibilidad/tratamiento farmacológico , Trastornos por Fotosensibilidad/genética , Trastornos por Fotosensibilidad/metabolismo , Protoporfiria Eritropoyética/inducido químicamente , Protoporfiria Eritropoyética/tratamiento farmacológico , Protoporfiria Eritropoyética/genética , Protoporfiria Eritropoyética/metabolismo , Protoporfirinas/genética , Protoporfirinas/metabolismo
10.
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
11.
Cells ; 8(7)2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31295943

RESUMEN

Aberrant activation of signaling pathways is frequently observed and reported to be associated with the progression and poor prognosis of prostate cancer (PCa). We aimed to identify key biological processes regulated by androgen receptor (AR) using gene co-expression network from single cell resolution. The bimodal index was used to evaluate whether two subpopulations exist among the single cells. Gene expression among single cells revealed averaging pitfalls and bimodality pattern. Weighted gene co-expression network analysis (WGCNA) was used to identify modules of highly correlated genes. Twenty-nine gene modules were identified and AR-regulated modules were screened by significantly overlapping reported androgen induced differentially expressed genes. The biological function "generation of precursor metabolites and energy" was significantly enriched by AR-regulated modules with bimodality, presenting differential androgen response among subpopulations. Integrating with public ChIP-seq data, two genes FECH, and CROT has AR binding sites. Public in vitro studies also show that androgen regulates FECH and CROT. After receiving androgen deprivation therapy, patients lowly express FECH and CROT. Further survival analysis indicates that FECH/CROT signature can predict PCa recurrence. We reveal the heterogeneous function of "generation of precursor metabolites and energy" upon androgen stimulation from the perspective of single cells. Inhibitors targeting this biological process will facilitate to prevent prostate cancer progression.


Asunto(s)
Carnitina Aciltransferasas/genética , Ferroquelatasa/genética , Neoplasias de la Próstata/genética , Antagonistas de Andrógenos , Andrógenos , Carnitina Aciltransferasas/metabolismo , Línea Celular Tumoral , Bases de Datos Genéticas , Ferroquelatasa/metabolismo , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Humanos , Masculino , Recurrencia Local de Neoplasia/genética , Pronóstico , Receptores Androgénicos/biosíntesis , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal/genética , Análisis de la Célula Individual/métodos , Transcriptoma/genética
12.
Mol Genet Metab ; 128(3): 304-308, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31076252

RESUMEN

Deficiency in ferrochelatase (FECH), the last enzyme in the heme biosynthetic pathway, leads to an accumulation of protoporphyrin IX (PPIX) that causes a severely painful phototoxic reaction of the skin in patients with erythropoietic protoporphyria (EPP). Besides phototoxicity of the skin, EPP patients often present with symptoms of iron deficiency in form of a microcytic and hypochromic anemia with low serum iron and ferritin. In addition, elevated aminolevulinic acid synthase 2 (ALAS2) both at the mRNA and protein levels have been observed among EPP patients. ALAS is the first enzyme in the pathway and exists in two isoforms, whereby the isoform 2 (ALAS2) is expressed exclusively in erythropoiesis. The mRNA of ALAS2 contains an iron response element (IRE) at its 5'UTR. When iron is limited, iron response element binding protein 2 (IRP2) binds to the IRE of ALAS2 mRNA and suppresses its translation. In this study, we demonstrated that iron deprivation increased the amount of ALAS2 mRNA as well as the ratio of ALAS2 to FECH mRNAs in cultured erythroleukemic K562 cells. At the protein level, however, iron deprivation in the cell line caused reductions in both enzymes as shown by the Western blot analysis. A comparable increase in the ratio of ALAS2 to FECH mRNAs was also found in EPP patients indicating an imbalance in heme biosynthesis. As iron cannot be completely missing from an organism, we assume that in EPP patients, a certain amount of ALAS2 mRNA is translated despite a partial deficiency of FECH. The increase in ALAS2 enzyme contributes to the accumulation in PPIX in the patients. Targeted inhibition of ALAS2 could therefore be a treatment option for EPP.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , 5-Aminolevulinato Sintetasa/metabolismo , Hierro/metabolismo , Protoporfiria Eritropoyética/enzimología , Vías Biosintéticas , Ferroquelatasa/genética , Humanos , Hierro/sangre , Proteína 2 Reguladora de Hierro/metabolismo , Proteínas Reguladoras del Hierro/metabolismo , Células K562 , Protoporfiria Eritropoyética/terapia , Protoporfirinas/metabolismo
13.
Photochem Photobiol ; 95(4): 1052-1059, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30767226

RESUMEN

Aminolevulinic acid (ALA) is a prodrug that is metabolized in the heme biosynthesis pathway to produce protoporphyrin IX (PpIX) for tumor fluorescence detection and photodynamic therapy (PDT). The iron chelator deferoxamine (DFO) has been widely used to enhance PpIX accumulation by inhibiting the iron-dependent bioconversion of PpIX to heme, a reaction catalyzed by ferrochelatase (FECH). Tumor response to DFO treatment is known to be highly variable, and some tumors even show no response. Given the fact that tumors often exhibit reduced FECH expression/enzymatic activity, we examined how reducing FECH level affected the DFO enhancement effect. Our results showed that reducing FECH level by silencing FECH in SkBr3 breast cancer cells completely abrogated the enhancement effect of DFO. Although DFO enhanced ALA-PpIX fluorescence and PDT response in SkBr3 vector control cells, it caused a similar increase in MCF10A breast epithelial cells, resulting in no net gain in the selectivity toward tumor cells. We also found that DFO treatment induced less increase in ALA-PpIX fluorescence in tumor cells with lower FECH activity (MDA-MB-231, Hs 578T) than in tumor cells with higher FECH activity (MDA-MB-453). Our study demonstrates that FECH activity is an important determinant of tumor response to DFO treatment.


Asunto(s)
Ácido Aminolevulínico/farmacología , Deferoxamina/farmacología , Ferroquelatasa/metabolismo , Fármacos Fotosensibilizantes/farmacología , Protoporfirinas/farmacología , Ácido Aminolevulínico/administración & dosificación , Neoplasias de la Mama , Línea Celular Tumoral , Células Epiteliales , Femenino , Ferroquelatasa/genética , Silenciador del Gen , Humanos , Fotoquimioterapia/métodos , Fármacos Fotosensibilizantes/administración & dosificación , Protoporfirinas/administración & dosificación , Sideróforos/farmacología
14.
World J Gastroenterol ; 25(7): 880-887, 2019 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-30809087

RESUMEN

BACKGROUND: Porphyria is a rare disease with complex classification. Erythropoietic protoporphyria (EPP) is an autosomal recessively inherited disease, and most are caused by mutations in the FECH gene. EPP combined with liver injury is even rarer. CASE SUMMARY: This paper reports a case of EPP which was admitted to the hospital with abnormal liver function and diagnosed by repeated questioning of medical history, screening of common causes of severe liver injury, and second generation sequencing of the whole exon genome. We also summarize the clinical characteristics of EPP with liver injury, and put forward some suggestions on EPP to provide a reference for the diagnosis of such rare disease. CONCLUSION: A new mutation locus (c.32_35dupCCCT) which may be related to the disease was found by detecting the FECH gene in the pedigree of this case.


Asunto(s)
Ferroquelatasa/genética , Hepatitis/diagnóstico , Protoporfiria Eritropoyética/diagnóstico , Enfermedades Raras/diagnóstico , Adulto , Biopsia , Análisis Mutacional de ADN , Errores Diagnósticos , Hepatitis/etiología , Hepatitis/patología , Humanos , Hígado/metabolismo , Hígado/patología , Pruebas de Función Hepática , Masculino , Linaje , Protoporfiria Eritropoyética/complicaciones , Protoporfiria Eritropoyética/genética , Protoporfiria Eritropoyética/patología , Enfermedades Raras/complicaciones , Piel/patología
15.
Mol Genet Metab ; 128(3): 342-351, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30660387

RESUMEN

Non-syndromic microcytic congenital sideroblastic anemia (cSA) is predominantly caused by defective genes encoding for either ALAS2, the first enzyme of heme biosynthesis pathway or SLC25A38, the mitochondrial importer of glycine, an ALAS2 substrate. Herein we explored a new case of cSA with two mutations in GLRX5, a gene for which only two patients have been reported so far. The patient was a young female with biallelic compound heterozygous mutations in GLRX5 (p.Cys67Tyr and p.Met128Lys). Three-D structure analysis confirmed the involvement of Cys67 in the coordination of the [2Fe2S] cluster and suggested a potential role of Met128 in partner interactions. The protein-level of ferrochelatase, the terminal-enzyme of heme process, was increased both in patient-derived lymphoblastoid and CD34+ cells, however, its activity was drastically decreased. The activity of ALAS2 was found altered and possibly related to a defect in the biogenesis of its co-substrate, the succinyl-CoA. Thus, the patient exhibits both a very low ferrochelatase activity without any accumulation of porphyrins precursors in contrast to what is reported in erythropoietic protoporphyria with solely impaired ferrochelatase activity. A significant oxidative stress was evidenced by decreased reduced glutathione and aconitase activity, and increased MnSOD protein expression. This oxidative stress depleted and damaged mtDNA, decreased complex I and IV activities and depleted ATP content. Collectively, our study demonstrates the key role of GLRX5 in modulating ALAS2 and ferrochelatase activities and in maintaining mitochondrial function.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/genética , Ferroquelatasa/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Glutarredoxinas/genética , Hemo/biosíntesis , Mutación Missense , 5-Aminolevulinato Sintetasa/metabolismo , Aconitato Hidratasa/metabolismo , Adolescente , Secuencia de Aminoácidos , Anemia Sideroblástica/enzimología , Línea Celular Transformada , Femenino , Ferroquelatasa/metabolismo , Enfermedades Genéticas Ligadas al Cromosoma X/enzimología , Glutatión/metabolismo , Humanos , Mitocondrias/enzimología , Estrés Oxidativo , Linaje , Estructura Terciaria de Proteína
16.
Mar Biotechnol (NY) ; 20(6): 829-843, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30145744

RESUMEN

With the emergence of several infectious diseases in shrimp aquaculture, there is a growing interest in the use of feed additives to enhance shrimp immunity. Recently, the use of 5-aminolevulinic acid (5-ALA), a non-protein amino acid that plays a rate-limiting role in heme biosynthesis, has received attention for its positive effect on immunity in livestock animals. To evaluate the effect of 5-ALA in the Pacific white shrimp, Litopenaeus vannamei, we conducted microarray analysis, a Vibrio parahaemolyticus immersion challenge test, an ATP level assay, and gene expression analysis of some hemoproteins and genes associated with heme synthesis and degradation. Out of 15,745 L. vannamei putative genes on the microarray, 101 genes were differentially expressed by more than fourfold (p < 0.05) between 5-ALA-supplemented and control shrimp hepatopancreas. 5-ALA upregulated 99 of the 101 genes, 41 of which were immune- and defense-related genes based on sequence homology. Compared to the control, the 5-ALA-supplemented group had a higher survival rate in the challenge test, higher transcript levels of porphobilinogen synthase, ferrochelatase, catalase, nuclear receptor E75, and heme oxygenase-1 and higher levels of ATP. These findings suggest that dietary 5-ALA enhanced the immune response of L. vannamei to V. parahaemolyticus, upregulated immune- and defense-related genes, and enhanced aerobic energy metabolism, respectively. Further studies are needed to elucidate the extent of 5-ALA use in shrimp culture.


Asunto(s)
Adenosina Trifosfato/metabolismo , Ácido Aminolevulínico/farmacología , Penaeidae/inmunología , Penaeidae/metabolismo , Animales , Catalasa/genética , Ferroquelatasa/genética , Hemo/metabolismo , Hemo-Oxigenasa 1/genética , Inmunidad/efectos de los fármacos , Penaeidae/efectos de los fármacos , Porfobilinógeno Sintasa/genética
17.
Mol Microbiol ; 109(3): 385-400, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29989674

RESUMEN

Haem is an essential cofactor in central metabolic pathways in the vast majority of living systems. Prokaryotes acquire haem via haem biosynthesis pathways, and some also utilize haem uptake systems, yet it remains unclear how they balance haem requirements with the paradox that free haem is toxic. Here, using the model pathogen Staphylococcus aureus, we report that IsdG, one of two haem oxygenase enzymes in the haem uptake system, inhibits the formation of haem via the internal haem biosynthesis route. More specifically, we show that IsdG decreases the activity of ferrochelatase and that the two proteins interact both in vitro and in vivo. Further, a bioinformatics analysis reveals that a significant number of haem biosynthesis pathway containing organisms possess an IsdG-homologue and that those with both biosynthesis and uptake systems have at least two haem oxygenases. We conclude that IsdG-like proteins control intracellular haem levels by coupling the two pathways. IsdG is thus a target for the treatment of S. aureusinfections.


Asunto(s)
Hemo/biosíntesis , Oxigenasas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/enzimología , Animales , Línea Celular , Ferroquelatasa/genética , Ferroquelatasa/metabolismo , Genes Bacterianos/genética , Humanos , Hierro/metabolismo , Macrófagos/microbiología , Ratones , Oxigenasas/genética , ARN Bacteriano/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Staphylococcus aureus/genética
18.
BMC Plant Biol ; 17(1): 187, 2017 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-29084526

RESUMEN

BACKGROUND: Non-essential trance metal such as cadmium (Cd) is toxic to plants. Although some plants have developed elaborate strategies to deal with absorbed Cd through multiple pathways, the regulatory mechanisms behind the Cd tolerance are not fully understood. Ferrochelatase-1 (FC1, EC4.99.1.1) is the terminal enzyme of heme biosynthesis, catalyzing insertion of ferrous ion into protoporphyrin IX. Recent studies have shown that FC1 is involved in several physiological processes. However, its biological function associated with plant abiotic stress response is poorly understood. RESULTS: In this study, we showed that AtFC1 was transcriptionally activated by Cd exposure. AtFC1 overexpression (35S::FC1) lines accumulated more Cd and non-protein thiol compounds than wild-type, and conferred plant tolerance to Cd stress, with improved primary root elongation, biomass and chlorophyll (Chl) content, and low degree of oxidation associated with reduced H2O2, O·2- and peroxides. In contrast, the AtFC1 loss of functional mutant fc1 showed sensitivity to Cd stress. Exogenous provision of heme, the product of AtFC1, partially rescued the Cd-induced toxic phenotype of fc1 mutants by improving the growth of seedlings, generation of glutathione (GSH) and phytochelatins (PCs), and GSH/PCs-synthesized gene expression (e.g. GSH1, GSH2, PCS1, and PCS2). To investigate the mechanism leading to the AtFC1 regulating Cd stress response in Arabidopsis, a transcriptome of fc1 mutant plants under Cd stress was profiled. Our data showed that disfunction of AtFC1 led to 913 genes specifically up-regulated and 522 genes down-regulated in fc1 mutants exposed to Cd. Some of the genes are involved in metal transporters, Cd-induced oxidative stress response, and detoxification. CONCLUSION: These results indicate that AtFC1 would act as a positive regulator of plant tolerance to Cd stress. Our study will broaden our understanding of the role of FC1 in mediating plant response to Cd stress and provide a basis for further exploration of its downstream genes.


Asunto(s)
Proteínas de Arabidopsis/genética , Cadmio/toxicidad , Ferroquelatasa/genética , Genes de Plantas/fisiología , Arabidopsis/enzimología , Arabidopsis/genética , Arabidopsis/fisiología , Proteínas de Arabidopsis/fisiología , Ferroquelatasa/fisiología , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Genes de Plantas/genética , Estrés Fisiológico/genética
19.
Mol Microbiol ; 106(6): 961-975, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29030914

RESUMEN

Facultative phototrophs such as Rhodobacter sphaeroides can switch between heterotrophic and photosynthetic growth. This transition is governed by oxygen tension and involves the large-scale production of bacteriochlorophyll, which shares a biosynthetic pathway with haem up to protoporphyrin IX. Here, the pathways diverge with the insertion of Fe2+ or Mg2+ into protoporphyrin by ferrochelatase or magnesium chelatase, respectively. Tight regulation of this branchpoint is essential, but the mechanisms for switching between respiratory and photosynthetic growth are poorly understood. We show that PufQ governs the haem/bacteriochlorophyll switch; pufQ is found within the oxygen-regulated pufQBALMX operon encoding the reaction centre-light-harvesting photosystem complex. A pufQ deletion strain synthesises low levels of bacteriochlorophyll and accumulates the biosynthetic precursor coproporphyrinogen III; a suppressor mutant of this strain harbours a mutation in the hemH gene encoding ferrochelatase, substantially reducing ferrochelatase activity and increasing cellular bacteriochlorophyll levels. FLAG-immunoprecipitation experiments retrieve a ferrochelatase-PufQ-carotenoid complex, proposed to regulate the haem/bacteriochlorophyll branchpoint by directing porphyrin flux toward bacteriochlorophyll production under oxygen-limiting conditions. The co-location of pufQ and the photosystem genes in the same operon ensures that switching of tetrapyrrole metabolism toward bacteriochlorophyll is coordinated with the production of reaction centre and light-harvesting polypeptides.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacterioclorofilas/metabolismo , Ferroquelatasa/metabolismo , Procesos Heterotróficos , Complejos de Proteína Captadores de Luz/metabolismo , Proteínas del Complejo del Centro de Reacción Fotosintética/metabolismo , Procesos Fototróficos , Rhodobacter sphaeroides/metabolismo , Aerobiosis , Anaerobiosis , Proteínas Bacterianas/genética , Carotenoides/metabolismo , Coproporfirinógenos/metabolismo , Ferroquelatasa/genética , Hemo/metabolismo , Complejos de Proteína Captadores de Luz/genética , Liasas/metabolismo , Mutación , Operón , Proteínas del Complejo del Centro de Reacción Fotosintética/genética , Protoporfirinas/metabolismo , Rhodobacter sphaeroides/genética , Tetrapirroles/biosíntesis
20.
Clin Chem ; 63(10): 1614-1623, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28784691

RESUMEN

BACKGROUND: There is much interest in the tissue of origin of circulating DNA in plasma. Data generated using DNA methylation markers have suggested that hematopoietic cells of white cell lineages are important contributors to the circulating DNA pool. However, it is not known whether cells of the erythroid lineage would also release DNA into the plasma. METHODS: Using high-resolution methylation profiles of erythroblasts and other tissue types, 3 genomic loci were found to be hypomethylated in erythroblasts but hypermethylated in other cell types. We developed digital PCR assays for measuring erythroid DNA using the differentially methylated region for each locus. RESULTS: Based on the methylation marker in the ferrochelatase gene, erythroid DNA represented a median of 30.1% of the plasma DNA of healthy subjects. In subjects with anemia of different etiologies, quantitative analysis of circulating erythroid DNA could reflect the erythropoietic activity in the bone marrow. For patients with reduced erythropoietic activity, as exemplified by aplastic anemia, the percentage of circulating erythroid DNA was decreased. For patients with increased but ineffective erythropoiesis, as exemplified by ß-thalassemia major, the percentage was increased. In addition, the plasma concentration of erythroid DNA was found to correlate with treatment response in aplastic anemia and iron deficiency anemia. Plasma DNA analysis using digital PCR assays targeting the other 2 differentially methylated regions showed similar findings. CONCLUSIONS: Erythroid DNA is a hitherto unrecognized major component of the circulating DNA pool and is a noninvasive biomarker for differential diagnosis and monitoring of anemia.


Asunto(s)
Anemia/sangre , Anemia/genética , Metilación de ADN , ADN/sangre , ADN/genética , Eritroblastos/patología , Anemia/diagnóstico , Anemia/patología , Anemia Aplásica/sangre , Anemia Aplásica/diagnóstico , Anemia Aplásica/genética , Anemia Aplásica/patología , Anemia Ferropénica/sangre , Anemia Ferropénica/diagnóstico , Anemia Ferropénica/genética , Anemia Ferropénica/patología , Diagnóstico Diferencial , Eritroblastos/metabolismo , Eritropoyesis , Ferroquelatasa/genética , Humanos , Síndromes Mielodisplásicos/sangre , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Talasemia beta/sangre , Talasemia beta/diagnóstico , Talasemia beta/genética , Talasemia beta/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA