Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Dis Esophagus ; 14(2): 162-5, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11553230

RESUMEN

Metastasis to the breast from extramammary malignancies is rare. This is the third case report of metastatic breast cancer from esophageal cancer. We report the clinical, radiographic, and pathologic findings of a 57-year-old woman who underwent esophagectomy for esophageal cancer and developed metastatic cancer 2 years later. Pathologic examination of a resected specimen of the breast revealed squamous cell carcinoma invading the mammary glands. Estrogen receptor and axillary lymph node metastasis were negative with immunostaining. She is alive 6 months after the modified radical mastectomy.


Asunto(s)
Neoplasias de la Mama/secundario , Neoplasias Esofágicas/patología , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/terapia , Terapia Combinada , Neoplasias Esofágicas/radioterapia , Neoplasias Esofágicas/cirugía , Femenino , Humanos , Persona de Mediana Edad
3.
Ryoikibetsu Shokogun Shirizu ; (33): 184-6, 2001.
Artículo en Japonés | MEDLINE | ID: mdl-11462392
4.
Blood ; 97(10): 2972-8, 2001 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-11342419

RESUMEN

The properties of 9 delta-aminolevulinate dehydratase (ALAD) mutants from patients with ALAD porphyria (ADP) were examined by bacterial expression of their complementary DNAs and by enzymologic and immunologic assays. ALADs were expressed as glutathione-S-transferase (GST) fusion proteins in Escherichia coli and purified by glutathione-affinity column chromatography. The GST-ALAD fusion proteins were recognized by anti-ALAD antibodies and were enzymatically active as ALAD. The enzymatic activities of 3 ALAD mutants, K59N, A274T, and V153M, were 69.9%, 19.3%, and 41.0% of that of the wild-type ALAD, respectively, whereas 6 mutants, G133R, K59N/G133R, F12L, R240W, V275M, and delTC, showed little activity (< 8%). These variations generally reflect the phenotype of ALAD in vivo in patients with ADP and indicate that GST-ALAD fusion protein is indeed useful for predicting of the phenotype of ALAD mutants. The location of F12L mutation in the enzyme's molecular structure indicates that its disturbance of the quaternary contact of the ALAD dimer appears to have a significant influence on the enzymatic activity. Mouse monoclonal antibodies to human ALAD were developed that specifically recognized a carboxy terminal portion of ALAD, or other regions in the enzyme. This study represents the first complete analysis of 9 mutants of ALAD identified in ADP and indicates the highly heterogeneous nature of mutations in this disorder.


Asunto(s)
Mutación , Porfobilinógeno Sintasa/deficiencia , Porfobilinógeno Sintasa/genética , Porfirias/enzimología , Adolescente , Animales , Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos , Escherichia coli/genética , Femenino , Expresión Génica , Glutatión Transferasa/genética , Humanos , Immunoblotting , Recién Nacido , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , Fenotipo , Porfobilinógeno Sintasa/metabolismo , Porfirias/genética , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo
5.
Eur J Biochem ; 268(6): 1705-11, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11248690

RESUMEN

Coproporphyrinogen oxidase (CPOX), the sixth enzyme in the heme-biosynthetic pathway, catalyzes oxidative decarboxylation of coproporphyrinogen to protoporphyrinogen and is located in the intermembrane space of mitochondria. To clarify the importance of CPOX in the regulation of heme biosynthesis in erythroid cells, we established human erythroleukemia K562 cells stably expressing mouse CPOX. The CPOX cDNA-transfected cells had sevenfold higher CPOX activity than cells transfected with vector only. Expression of ferrochelatase and heme content in the transfected cells increased slightly compared with the control. When K562 cells overexpressing CPOX were treated with delta-aminolevulinic acid (ALA), most became benzidine-positive without induction of the expression of CPOX or ferrochelatase, and the heme content was about twofold higher than that in ALA-treated control cells. Increases in cellular heme concomitant with a marked induction of the expression of heme-biosynthetic enzymes, including CPOX, ferrochelatase and erythroid-specific delta-aminolevulinic acid synthase, as well as of alpha-globin synthesis, were observed when cells were treated with transforming growth factor (TGF)beta 1. These increases in the transfected cells were twice those in control cells, indicating that overexpression of CPOX enhanced induction of the differentiation of K562 cells mediated by TGF beta 1 or ALA. Conversely, the transfection of antisense oligonucleotide to human CPOX mRNA into untreated and TGF beta 1-treated K562 cells led to a decrease in heme production compared with sense oligonucleotide-transfected cells. These results suggest that CPOX plays an important role in the regulation of heme biosynthesis during erythroid differentiation.


Asunto(s)
Coproporfirinógeno Oxidasa/metabolismo , Hemoglobinas/biosíntesis , Leucemia Eritroblástica Aguda/enzimología , Ácido Aminolevulínico/farmacología , Animales , Secuencia de Bases , Coproporfirinógeno Oxidasa/genética , Cartilla de ADN , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Hemoglobinas/genética , Humanos , Células K562 , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patología , Ratones , Oligonucleótidos Antisentido/farmacología , Transfección , Factor de Crecimiento Transformador beta/farmacología
7.
Biochem Biophys Res Commun ; 273(2): 596-602, 2000 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-10873651

RESUMEN

Coproporphyrinogen oxidase [CPO] gene promoter regulatory element (CPRE) plays an important role in CPO gene regulation. To isolate a CPRE binding protein, we performed Southwestern screening of K562 cDNA expression library using CPRE as a probe and isolated a cDNA clone which encoded a novel protein, Klp1 (K562 cell-derived leucine-zipper-like protein 1). Klp1 mRNA was highly expressed in K562 cells, HeLa cells, and brain as a single transcript (1.4 kb). Gel mobility shift assays revealed that Klp1 specifically binds to CPRE. Computational analysis revealed that Klp1 has a leucine-zipper-like structure, a Leu-X-X-Leu-Leu motif, and a putative nuclear localization signal in the basic amino acid rich region. Transfection of the Klp1 expression vector into THP-1 cells resulted in transcriptional activation of a reporter construct containing CPRE. These results indicate that Klp1 is a DNA sequence-specific transcription factor that regulates gene expression of genes that contain CPRE in their regulatory region.


Asunto(s)
Coproporfirinógeno Oxidasa/genética , Proteínas de Unión al ADN/genética , Regiones Promotoras Genéticas , Transactivadores/genética , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/genética , Línea Celular , Clonación Molecular , Cartilla de ADN/genética , ADN Complementario/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Expresión Génica , Células HeLa , Humanos , Células K562 , Datos de Secuencia Molecular , Embarazo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Distribución Tisular , Transactivadores/metabolismo , Células U937
8.
J Clin Invest ; 105(6): 757-64, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10727444

RESUMEN

The first and the rate-limiting enzyme of heme biosynthesis is delta-aminolevulinate synthase (ALAS), which is localized in mitochondria. There are 2 tissue-specific isoforms of ALAS, erythroid-specific (ALAS-E) and nonspecific ALAS (ALAS-N). To identify possible mitochondrial factors that modulate ALAS-E function, we screened a human bone marrow cDNA library, using the mitochondrial form of human ALAS-E as a bait protein in the yeast 2-hybrid system. Our screening led to the isolation of the beta subunit of human ATP-specific succinyl CoA synthetase (SCS-betaA). Using transient expression and coimmunoprecipitation, we verified that mitochodrially expressed SCS-betaA associates specifically with ALAS-E and not with ALAS-N. Furthermore, the ALAS-E mutants R411C and M426V associated with SCS-betaA, but the D190V mutant did not. Because the D190V mutant was identified in a patient with pyridoxine-refractory X-linked sideroblastic anemia, our findings suggest that appropriate association of SCS-betaA and ALAS-E promotes efficient use of succinyl CoA by ALAS-E or helps translocate ALAS-E into mitochondria.


Asunto(s)
5-Aminolevulinato Sintetasa/metabolismo , Anemia Sideroblástica/enzimología , Hemo/biosíntesis , Isoenzimas/metabolismo , Succinato-CoA Ligasas/metabolismo , 5-Aminolevulinato Sintetasa/genética , Acilcoenzima A/metabolismo , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Médula Ósea/química , Células CHO , Cricetinae , Cricetulus , ADN Complementario/genética , Inducción Enzimática , Células Precursoras Eritroides/enzimología , Humanos , Isoenzimas/genética , Sustancias Macromoleculares , Mitocondrias Cardíacas/enzimología , Datos de Secuencia Molecular , Miocardio/química , Mutación Puntual , ARN Mensajero/biosíntesis , Técnicas del Sistema de Dos Híbridos
9.
Hepatology ; 31(3): 704-8, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10706561

RESUMEN

Cloning and expression of the defective gene for delta-aminolevulinate dehydratase (ALAD) from the second of 2 German patients with ALAD deficiency porphyria (ADP), who had been originally reported by Doss et al. in 1979, were performed. Cloning of cDNAs for the defective ALAD were performed using Epstein-Barr virus (EBV)-transformed lymphoblastoid cells of the proband, and nucleotide sequences of cloned cDNA were determined. Two separate mutations of ALAD cDNA were identified in each ALAD allele. One was G457A, termed "H1," resulting in V153M substitution, while the other was a deletion of 2 sequential bases at T(818) and C(819), termed "H2," resulting in a frame shift with a premature stop codon at the amino acid position of 294. Using allele-specific oligonucleotide hybridization, the mother of the proband was shown to have an H1 defect, while using genomic DNA analysis, the father was shown to have an H2 defect. Expression of H1 cDNA in Chinese hamster ovary cells produced an ALAD protein with only a partial activity (10.65% +/- 1.80% of the normal), while H2 cDNA encoded no significant protein. These data thus demonstrate that the proband was associated with 2 novel molecular defects of the ALAD gene, 1 in each allele, and account for the extremely low ALAD activity in his erythrocytes ( approximately 1% of normal).


Asunto(s)
Mutación , Porfobilinógeno Sintasa/genética , Porfirias Hepáticas/genética , Enfermedad Aguda , Adolescente , Animales , Western Blotting , Células CHO , Clonación Molecular , Cricetinae , ADN Complementario/biosíntesis , Eritrocitos/enzimología , Humanos , Masculino , Porfobilinógeno Sintasa/deficiencia , Porfirias Hepáticas/sangre , Porfirias Hepáticas/enzimología
11.
EMBO J ; 18(22): 6282-9, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10562540

RESUMEN

Erythroid 5-aminolevulinate synthase (ALAS-E) catalyzes the first step of heme biosynthesis in erythroid cells. Mutation of human ALAS-E causes the disorder X-linked sideroblastic anemia. To examine the roles of heme during hematopoiesis, we disrupted the mouse ALAS-E gene. ALAS-E-null embryos showed no hemoglobinized cells and died by embryonic day 11.5, indicating that ALAS-E is the principal isozyme contributing to erythroid heme biosynthesis. In the ALAS-E-null mutant embryos, erythroid differentiation was arrested, and an abnormal hematopoietic cell fraction emerged that accumulated a large amount of iron diffusely in the cytoplasm. In contrast, we found typical ring sideroblasts that accumulated iron mostly in mitochondria in adult mice chimeric for ALAS-E-null mutant cells, indicating that the mode of iron accumulation caused by the lack of ALAS-E is different in primitive and definitive erythroid cells. These results demonstrate that ALAS-E, and hence heme supply, is necessary for differentiation and iron metabolism of erythroid cells.


Asunto(s)
5-Aminolevulinato Sintetasa/metabolismo , Células Precursoras Eritroides/citología , Eritropoyesis/genética , Hemo/deficiencia , Hierro/metabolismo , 5-Aminolevulinato Sintetasa/deficiencia , 5-Aminolevulinato Sintetasa/genética , Animales , Diferenciación Celular , Quimera , Citoplasma/metabolismo , Citoplasma/ultraestructura , Desarrollo Embrionario y Fetal , Células Precursoras Eritroides/ultraestructura , Femenino , Muerte Fetal , Citometría de Flujo , Genotipo , Humanos , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Noqueados
12.
Am J Hematol ; 62(2): 112-4, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10577279

RESUMEN

A novel missense mutation, G663A, in exon 5 of the erythroid-specific delta-aminolevulinate synthase gene (ALAS2) was identified in a Japanese male with pyridoxine-responsive sideroblastic anemia. Activity of the mutant delta-aminolevulinate synthase protein expressed in vitro was 15.1% compared with the normal control, but was increased up to 34.5% by the addition of pyridoxal 5'-phosphate, consistent with the clinical response of the patient to pyridoxine treatment. The same mutation was also detected in genomic DNa from the oral mucosal membrane of the patiet; however, it was not detected in other family member. These findings suggest that this G663A mutation is responsible for sideroblastic anemia in the proband, and may be an index mutation in this pedigree.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/sangre , Anemia Sideroblástica/genética , Eritrocitos/enzimología , Mutación Missense , Piridoxina/uso terapéutico , 5-Aminolevulinato Sintetasa/sangre , Anemia Sideroblástica/tratamiento farmacológico , Anemia Sideroblástica/enzimología , Secuencia de Bases , Células de la Médula Ósea/patología , Exones , Humanos , Lactante , Japón , Masculino , Reacción en Cadena de la Polimerasa , Valores de Referencia
13.
Rinsho Ketsueki ; 40(7): 593-8, 1999 Jul.
Artículo en Japonés | MEDLINE | ID: mdl-10483144

RESUMEN

A 20-year-old man presented with microcytic hypochromic anemia (hemoglobin: 9.3 g/dl, MCV: 82.0 fl, MCHC: 29.5 g/dl) and dimorphism RBCs in circulating blood (RDW: 26.8%). Ringed sideroblasts accounted for 29.5% of bone marrow erythroblasts. Iron overload was also observed. Because the patient had a clear family history of anemia, he was given a diagnosis of X-linked sideroblastic anemia. The activity of delta-aminolevulinic acid synthase (ALAS) in bone marrow erythroblasts was low. However, we did not detect mutation of the gene for ALAS. The patient has responded well to a treatment regimen consisting of oral vitamin B6, Fe-chelation therapy, and phlebotomy.


Asunto(s)
Anemia Sideroblástica/genética , Ligamiento Genético , Cromosoma X , Adulto , Humanos , Masculino , Linaje
14.
Br J Haematol ; 106(1): 175-7, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10444183

RESUMEN

A novel missense mutation, A1754G, in exon 11 of the erythroid-specific delta-aminolaevulinate synthase gene (ALAS2) was identified in a Japanese male with sideroblastic anaemia. ALAS activity in bone marrow cells of the patient was reduced to 53.3% of the normal control. Consistent with this finding, activity of a bacterially expressed ALAS2 mutant protein harbouring this mutation was 19.5% compared with the normal control, but was increased up to 31.6% by the addition of pyridoxal 5'-phosphate (PLP) in vitro. RFLP analysis with Bsp HI restriction revealed that his mother was a carrier of the mutation. These findings suggest that A1754G mutation was inherited in this family in a manner consistent with X-linked inheritance, and is responsible for sideroblastic anaemia in the patient.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/genética , Mutación Missense/genética , Adolescente , Humanos , Masculino , Polimorfismo de Longitud del Fragmento de Restricción
16.
FEBS Lett ; 439(1-2): 163-7, 1998 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-9849899

RESUMEN

Transcription levels of the non-specific delta-aminolevulinate synthase (ALAS-N) and heme oxygenase-1 (HO-1) in the placenta at the terminal stage of pregnancy were comparable to those in the female adult liver and in the spleen, respectively. Immunohistochemical studies demonstrated that both enzymes were exclusively expressed in the trophoblast. During gestation, transcript of ALAS-N slightly increased, while HO-1 mRNA significantly decreased. Induced acute fetal hypoxia resulted in an increase in ALAS-N mRNA and in a decrease in HO-1 mRNA. These findings indicate that placental heme metabolism is influenced by the oxygen supply.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Regulación del Desarrollo de la Expresión Génica , Hemo Oxigenasa (Desciclizante)/genética , Placenta/enzimología , 5-Aminolevulinato Sintetasa/metabolismo , Animales , Femenino , Hipoxia Fetal/enzimología , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1 , Inmunohistoquímica , Ligadura , Placenta/patología , Embarazo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Útero/irrigación sanguínea
17.
Br J Haematol ; 103(3): 839-41, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9858242

RESUMEN

A R411C missense mutation of the erythroid-specific delta-aminolaevulinate synthase (ALAS2) gene was identified in a pedigree with X-linked pyridoxine-responsive sideroblastic anaemia (XLSA). The normal and the mutant cDNAs were expressed in E. coli, and the enzyme protein was purified. ALAS activity of the mutant enzyme was 12% and 25%, when incubated in the absence and the presence of pyridoxal 5'-phosphate, respectively, compared with that of the wild-type enzyme. These findings suggest that the R411C mutation accounts for low ALAS activity and a partial pyridoxine-responsiveness of the disease in the patient.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/genética , Mutación Missense , Adolescente , Anemia Sideroblástica/tratamiento farmacológico , Anemia Sideroblástica/enzimología , Humanos , Masculino , Linaje , Piridoxina/uso terapéutico
19.
J Biol Chem ; 273(9): 5358-65, 1998 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-9478996

RESUMEN

The erythroid transcription factor NF-E2 is an obligate heterodimer composed of two different subunits (p45 and p18), each containing a basic region-leucine zipper DNA binding domain, and it plays a critical role in erythroid differentiation as an enhancer-binding protein for expression of the beta-globin gene. We show here that dimethyl sulfoxide treatment of wild-type murine erythroleukemia cells, but not a mutant clone of dimethyl sulfoxide-resistant cells, increases NF-E2 activity significantly, which involves both up-regulation of DNA binding and transactivation activities. Both activities were reduced markedly by treatment of cells with 2-aminopurine but not by genistein. Activation of the Ras-Raf-MAP kinase signaling cascade increased NF-E2 activity significantly, but this was suppressed when MafK was overexpressed. Domain analysis revealed an activation domain in the NH2-terminal region of p45 and a suppression domain in the basic region-leucine zipper of MafK. These findings indicate that induction of NF-E2 activity is essential for erythroid differentiation of murine erythroleukemia cells, and serine/threonine phosphorylation may be involved in this process. In addition, they also suggest that a MafK homodimer can suppress transcription, not only by competition for the DNA binding site, but also by direct inhibition of transcription. Hence, MafK may function as an active transcription repressor.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Leucemia Eritroblástica Aguda/genética , Factores de Transcripción/metabolismo , 5-Aminolevulinato Sintetasa/biosíntesis , Animales , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Diferenciación Celular , Dimetilsulfóxido/farmacología , Resistencia a Medicamentos/genética , Elementos de Facilitación Genéticos , Factores de Unión al ADN Específico de las Células Eritroides , Globinas/biosíntesis , Hemo/análisis , Factor de Transcripción MafK , Ratones , Modelos Genéticos , Factor de Transcripción NF-E2 , Subunidad p45 del Factor de Transcripción NF-E2 , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , ARN Mensajero/biosíntesis , Transducción de Señal , Activación Transcripcional , Dedos de Zinc , Proteínas ras/metabolismo
20.
Blood ; 90(2): 822-30, 1997 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-9226183

RESUMEN

To elucidate how pyridoxine-refractory X-linked sideroblastic anemia (XLSA) develops, we analyzed the erythroid-specific 5-aminolevulinate synthase (ALAS-E) gene of a patient with the anemia. The activity and amount of the enzyme in bone marrow cells of the patient were found to be approximately 5% of the normal control. We identified a point mutation, which introduces an amino acid substitution from Asp 190 to Val. In transient transfection analyses using quail fibroblasts, accumulation of aberrantly processed proteins, the sizes of which were larger than that of mature ALAS-E, was found in mitochondria. The proteins were reproducibly detected in assays combining in vitro transcription/translation of ALAS-E precursor and import of the precursor into isolated mouse mitochondria. These results suggest that the mutation causing pyridoxine-refractory XLSA affects the processing of the ALAS-E precursor, thus provoking instability of the ALAS-E protein.


Asunto(s)
5-Aminolevulinato Sintetasa/genética , Anemia Sideroblástica/enzimología , Anemia Sideroblástica/genética , Mutación Puntual , Piridoxina/uso terapéutico , Cromosoma X , 5-Aminolevulinato Sintetasa/biosíntesis , 5-Aminolevulinato Sintetasa/química , Secuencia de Aminoácidos , Anemia Sideroblástica/sangre , Animales , Ácido Aspártico , Secuencia de Bases , Médula Ósea/enzimología , Cartilla de ADN , Exones , Femenino , Biblioteca de Genes , Humanos , Intrones , Masculino , Ratones , Mitocondrias/enzimología , Biosíntesis de Proteínas , Ratas , Reproducibilidad de los Resultados , Transcripción Genética , Valina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA