Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 306
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nature ; 537(7618): 63-68, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27533040

RESUMEN

Hypermethylation of the promoters of tumour suppressor genes represses transcription of these genes, conferring growth advantages to cancer cells. How these changes arise is poorly understood. Here we show that the activity of oxygen-dependent ten-eleven translocation (TET) enzymes is reduced by tumour hypoxia in human and mouse cells. TET enzymes catalyse DNA demethylation through 5-methylcytosine oxidation. This reduction in activity occurs independently of hypoxia-associated alterations in TET expression, proliferation, metabolism, hypoxia-inducible factor activity or reactive oxygen species, and depends directly on oxygen shortage. Hypoxia-induced loss of TET activity increases hypermethylation at gene promoters in vitro. In patients, tumour suppressor gene promoters are markedly more methylated in hypoxic tumour tissue, independent of proliferation, stromal cell infiltration and tumour characteristics. Our data suggest that up to half of hypermethylation events are due to hypoxia, with these events conferring a selective advantage. Accordingly, increased hypoxia in mouse breast tumours increases hypermethylation, while restoration of tumour oxygenation abrogates this effect. Tumour hypoxia therefore acts as a novel regulator of DNA methylation.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/deficiencia , Oxigenasas de Función Mixta/deficiencia , Oxígeno/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Hipoxia Tumoral/fisiología , 5-Metilcitosina/metabolismo , Animales , Proliferación Celular , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Femenino , Silenciador del Gen/efectos de los fármacos , Genes Supresores de Tumor , Humanos , Masculino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Oxidación-Reducción/efectos de los fármacos , Oxígeno/farmacología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Células del Estroma/patología , Hipoxia Tumoral/efectos de los fármacos , Hipoxia Tumoral/genética
2.
Proc Natl Acad Sci U S A ; 116(32): 16036-16045, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31332008

RESUMEN

Cardiovascular disease (CVD) events due to atherosclerosis cause one-third of worldwide deaths and risk factors include physical inactivity, age, dyslipidemia, hypertension, diabetes, obesity, smoking, and red meat consumption. However, ∼15% of first-time events occur without such factors. In contrast, coronary events are extremely rare even in closely related chimpanzees in captivity, despite human-like CVD-risk-prone blood lipid profiles, hypertension, and mild atherosclerosis. Similarly, red meat-associated enhancement of CVD event risk does not seem to occur in other carnivorous mammals. Thus, heightened CVD risk may be intrinsic to humans, and genetic changes during our evolution need consideration. Humans exhibit a species-specific deficiency of the sialic acid N-glycolylneuraminic acid (Neu5Gc), due to pseudogenization of cytidine monophosphate-N-acetylneuraminic acid (Neu5Ac) hydroxylase (CMAH), which occurred in hominin ancestors ∼2 to 3 Mya. Ldlr-/- mice with human-like Cmah deficiency fed a sialic acids (Sias)-free high-fat diet (HFD) showed ∼1.9-fold increased atherogenesis over Cmah wild-type Ldlr-/- mice, associated with elevated macrophage cytokine expression and enhanced hyperglycemia. Human consumption of Neu5Gc (from red meat) acts as a "xeno-autoantigen" via metabolic incorporation into endogenous glycoconjugates, as interactions with circulating anti-Neu5Gc "xeno-autoantibodies" potentiate chronic inflammation ("xenosialitis"). Cmah-/-Ldlr-/- mice immunized with Neu5Gc-bearing antigens to generate human-like anti-Neu5Gc antibodies suffered a ∼2.4-fold increased atherosclerosis on a Neu5Gc-rich HFD, compared with Neu5Ac-rich or Sias-free HFD. Lesions in Neu5Gc-immunized and Neu5Gc-rich HFD-fed Cmah-/-Ldlr-/- mice were more advanced but unexplained by lipoprotein or glucose changes. Human evolutionary loss of CMAH likely contributes to atherosclerosis predisposition via multiple intrinsic and extrinsic mechanisms, and future studies could consider this more human-like model.


Asunto(s)
Aterosclerosis/enzimología , Oxigenasas de Función Mixta/deficiencia , Animales , Bovinos , Citocinas/metabolismo , Dieta Alta en Grasa , Femenino , Humanos , Hiperglucemia/patología , Inflamación/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Oxigenasas de Función Mixta/metabolismo , Modelos Biológicos , Fenotipo , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo , Ácidos Siálicos/metabolismo , Especificidad de la Especie
3.
Cytogenet Genome Res ; 160(9): 523-530, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33161406

RESUMEN

Sterol-C4-methyl oxidase (SC4MOL) deficiency was recently described as an autosomal recessive cholesterol biosynthesis disorder caused by mutations in the MSMO1 (sometimes also referred to as SC4MOL) gene. To date, 5 patients from 4 unrelated families with SC4MOL deficiency have been reported. Diagnosis can be challenging as the biochemical accumulation of methylsterols can affect global development and cause skin and ocular pathology. Herein, we describe 2 siblings from a consanguineous Turkish family with SC4MOL deficiency presenting with psoriasiform dermatitis, ocular abnormalities (nystagmus, optic hypoplasia, myopia, and strabismus), severe intellectual disability, and growth and motor delay. We undertook whole-exome sequencing and identified a new homozygous missense mutation c.81A>C; p.Asn27Thr in MSMO1. Segregation analysis in all available family members confirmed recessive inheritance of the mutation. The siblings were treated with a combination of oral and topical statin and cholesterol which resulted in clinical improvement. This study demonstrates how genomics-based diagnosis and therapy can be helpful in clinical practice.


Asunto(s)
Dermatitis/genética , Oxigenasas de Función Mixta/genética , Mutación Missense , Psoriasis/genética , Niño , Colesterol/biosíntesis , Colesterol/sangre , Colesterol/deficiencia , Colesterol/uso terapéutico , Consanguinidad , Oftalmopatías/genética , Femenino , Genes Recesivos , Trastornos del Crecimiento/genética , Homocigoto , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Discapacidad Intelectual/genética , Masculino , Oxigenasas de Función Mixta/deficiencia , Linaje , Rosuvastatina Cálcica/uso terapéutico , Hermanos , Triglicéridos/sangre , Turquía , Secuenciación del Exoma
4.
J Biol Chem ; 293(40): 15370-15380, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30126845

RESUMEN

Many aerobic organisms have developed molecular mechanism to tolerate hypoxia, but the specifics of these mechanisms remain poorly understood. It is important to develop genetic methods that confer increased hypoxia tolerance to intensively farmed aquatic species, as these are maintained in environments with limited available oxygen. As an asparaginyl hydroxylase of hypoxia-inducible factors (HIFs), factor inhibiting HIF (FIH) inhibits transcriptional activation of hypoxia-inducible genes by blocking the association of HIFs with the transcriptional coactivators CREB-binding protein (CBP) and p300. Therefore, here we sought to test whether fih is involved in regulating hypoxia tolerance in the commonly used zebrafish model. Overexpressing the zebrafish fih gene in epithelioma papulosum cyprini (EPC) cells and embryos, we found that fih inhibits the transcriptional activation of zebrafish HIF-α proteins. Using CRISPR/Cas9 to obtain fih-null zebrafish mutants, we noted that the fih deletion makes zebrafish more tolerant of hypoxic conditions than their WT siblings, but does not result in oxygen consumption rates that significantly differ from those of WT fish. Of note, we identified fewer apoptotic cells in adult fih-null zebrafish brains and in fih-null embryos, possibly explaining why the fih-null mutant had greater hypoxia tolerance than the WT. Moreover, the fih deletion up-regulated several hypoxia-inducible genes in fih-null zebrafish exposed to hypoxia. The findings of our study suggest that fih plays a role in hypoxia tolerance by affecting the rate of cellular apoptosis in zebrafish.


Asunto(s)
Adaptación Fisiológica/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Hipoxia/genética , Oxigenasas de Función Mixta/genética , Proteínas Represoras/genética , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Apoptosis/genética , Encéfalo/metabolismo , Encéfalo/fisiopatología , Proteína de Unión a CREB/genética , Proteína de Unión a CREB/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Embrión no Mamífero , Células Epiteliales/metabolismo , Células Epiteliales/patología , Eliminación de Gen , Edición Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Hipoxia/metabolismo , Hipoxia/fisiopatología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/metabolismo , Consumo de Oxígeno/genética , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/metabolismo
5.
Xenotransplantation ; 26(4): e12516, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30989742

RESUMEN

Xenotransplantation research has made considerable progress in recent years, largely through the increasing availability of pigs with multiple genetic modifications. We suggest that a pig with nine genetic modifications (ie, currently available) will provide organs (initially kidneys and hearts) that would function for a clinically valuable period of time, for example, >12 months, after transplantation into patients with end-stage organ failure. The national regulatory authorities, however, will likely require evidence, based on in vitro and/or in vivo experimental data, to justify the inclusion of each individual genetic modification in the pig. We provide data both from our own experience and that of others on the advantages of pigs in which (a) all three known carbohydrate xenoantigens have been deleted (triple-knockout pigs), (b) two human complement-regulatory proteins (CD46, CD55) and two human coagulation-regulatory proteins (thrombomodulin, endothelial cell protein C receptor) are expressed, (c) the anti-apoptotic and "anti-inflammatory" molecule, human hemeoxygenase-1 is expressed, and (d) human CD47 is expressed to suppress elements of the macrophage and T-cell responses. Although many alternative genetic modifications could be made to an organ-source pig, we suggest that the genetic manipulations we identify above will all contribute to the success of the initial clinical pig kidney or heart transplants, and that the beneficial contribution of each individual manipulation is supported by considerable experimental evidence.


Asunto(s)
Animales Modificados Genéticamente/genética , Rechazo de Injerto/prevención & control , Porcinos/genética , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente/inmunología , Antígeno CD47/genética , Antígeno CD47/inmunología , Antígenos CD55/genética , Antígenos CD55/inmunología , Receptor de Proteína C Endotelial/genética , Receptor de Proteína C Endotelial/inmunología , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Galactosiltransferasas/inmunología , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Rechazo de Injerto/inmunología , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/inmunología , Humanos , Proteína Cofactora de Membrana/genética , Proteína Cofactora de Membrana/inmunología , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/inmunología , N-Acetilgalactosaminiltransferasas/deficiencia , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/inmunología , Porcinos/inmunología , Trombomodulina/genética , Trombomodulina/inmunología
6.
Xenotransplantation ; 26(5): e12524, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31115108

RESUMEN

Two well-characterized carbohydrate epitopes are absent in humans but present in other mammals. These are galactose-α1,3-galactose (αGal) and N-glycolylneuraminic acid (Neu5Gc) which are introduced by the activities of two enzymes including α(1,3) galactosyltransferase (encoded by the GGTA1 gene) and CMP-Neu5Gc hydroxylase (encoded by the CMAH gene) that are inactive in humans but present in cattle. Hence, bovine-derived products are antigenic in humans who receive bioprosthetic heart valves (BHVs) or those that suffer from red meat syndrome. Using programmable nucleases, we disrupted (knockout, KO) GGTA1 and CMAH genes encoding for the enzymes that catalyse the synthesis of αGal and Neu5Gc, respectively, in both male and female bovine fibroblasts. The KO in clonally selected fibroblasts was detected by polymerase chain reaction (PCR) and confirmed by Sanger sequencing. Selected fibroblasts colonies were used for somatic cell nuclear transfer (SCNT) to produce cloned embryos that were implanted in surrogate recipient heifers. Fifty-three embryos were implanted in 33 recipients heifers; 3 pregnancies were carried to term and delivered 3 live calves. Primary cell cultures were established from the 3 calves and following molecular analyses confirmed the genetic deletions. FACS analysis showed the double-KO phenotype for both antigens confirming the mutated genotypes. Availability of such cattle double-KO model lacking both αGal and Neu5Gc offers a unique opportunity to study the functionality of BHV manufactured with tissues of potentially lower immunogenicity, as well as a possible new clinical approaches to help patients with red meat allergy syndrome due to the presence of these xenoantigens in the diet.


Asunto(s)
Animales Modificados Genéticamente , Antígenos Heterófilos/metabolismo , Citidina Monofosfato/análogos & derivados , Galactosa/metabolismo , Galactosiltransferasas/genética , Técnicas de Inactivación de Genes , Oxigenasas de Función Mixta/genética , Ácidos Neuramínicos/metabolismo , Animales , Antígenos Heterófilos/inmunología , Bioprótesis , Bovinos , Citidina Monofosfato/inmunología , Citidina Monofosfato/metabolismo , Femenino , Fibroblastos/inmunología , Hipersensibilidad a los Alimentos/inmunología , Galactosa/inmunología , Galactosiltransferasas/deficiencia , Prótesis Valvulares Cardíacas , Humanos , Masculino , Oxigenasas de Función Mixta/deficiencia , Ácidos Neuramínicos/inmunología , Trasplante Heterólogo
7.
Xenotransplantation ; 26(4): e12504, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30825348

RESUMEN

The humoral barrier has been the limiting factor in moving xenotransplantation towards the clinic. Improvements in somatic cell nuclear transfer and genome editing, particularly CRISPR-Cas9, have made it possible to create pigs with multiple glycan xenoantigen deletions for the purposes of reducing xenoreactive antibody binding to the xenografted organ. Recent studies have also considered the aetiology and existence of antibodies directed at the swine leucocyte antigen (SLA) complex, and potential genetic engineering strategies to avoid these antibodies. Evaluation of xenoreactive antibody binding is very important for the advancement of xenotransplantation, because if patients do not have any detectable xenoreactive antibody, then it is reasonable to expect that cellular rejection and not antibody-mediated rejection (AMR) will be the next hurdle to clinical application.


Asunto(s)
Antígenos Heterófilos/inmunología , Galactosiltransferasas/inmunología , Técnicas de Inactivación de Genes , Rechazo de Injerto/prevención & control , Oxigenasas de Función Mixta/inmunología , N-Acetilgalactosaminiltransferasas/inmunología , Porcinos/inmunología , Trasplante Heterólogo , Animales , Animales Modificados Genéticamente/inmunología , Anticuerpos Heterófilos/biosíntesis , Anticuerpos Heterófilos/inmunología , Reacciones Antígeno-Anticuerpo , Antígenos Heterófilos/genética , Epítopos/inmunología , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Ingeniería Genética , Rechazo de Injerto/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , N-Acetilgalactosaminiltransferasas/deficiencia , N-Acetilgalactosaminiltransferasas/genética , Inmunología del Trasplante
8.
Proc Biol Sci ; 285(1886)2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30209232

RESUMEN

Compared to other primates, humans are exceptional long-distance runners, a feature that emerged in genus Homo approximately 2 Ma and is classically attributed to anatomical and physiological adaptations such as an enlarged gluteus maximus and improved heat dissipation. However, no underlying genetic changes have currently been defined. Two to three million years ago, an exon deletion in the CMP-Neu5Ac hydroxylase (CMAH) gene also became fixed in our ancestral lineage. Cmah loss in mice exacerbates disease severity in multiple mouse models for muscular dystrophy, a finding only partially attributed to differences in immune reactivity. We evaluated the exercise capacity of Cmah-/- mice and observed an increased performance during forced treadmill testing and after 15 days of voluntary wheel running. Cmah-/- hindlimb muscle exhibited more capillaries and a greater fatigue resistance in situ Maximal coupled respiration was also higher in Cmah null mice ex vivo and relevant differences in metabolic pathways were also noted. Taken together, these data suggest that CMAH loss contributes to an improved skeletal muscle capacity for oxygen use. If translatable to humans, CMAH loss could have provided a selective advantage for ancestral Homo during the transition from forest dwelling to increased resource exploration and hunter/gatherer behaviour in the open savannah.


Asunto(s)
Ratones/fisiología , Oxigenasas de Función Mixta/metabolismo , Carrera , Animales , Masculino , Ratones/genética , Ratones Noqueados , Oxigenasas de Función Mixta/deficiencia , Condicionamiento Físico Animal
9.
Proc Natl Acad Sci U S A ; 112(2): 542-7, 2015 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-25548184

RESUMEN

A well known, epidemiologically reproducible risk factor for human carcinomas is the long-term consumption of "red meat" of mammalian origin. Although multiple theories have attempted to explain this human-specific association, none have been conclusively proven. We used an improved method to survey common foods for free and glycosidically bound forms of the nonhuman sialic acid N-glycolylneuraminic acid (Neu5Gc), showing that it is highly and selectively enriched in red meat. The bound form of Neu5Gc is bioavailable, undergoing metabolic incorporation into human tissues, despite being a foreign antigen. Interactions of this antigen with circulating anti-Neu5Gc antibodies could potentially incite inflammation. Indeed, when human-like Neu5Gc-deficient mice were fed bioavailable Neu5Gc and challenged with anti-Neu5Gc antibodies, they developed evidence of systemic inflammation. Such mice are already prone to develop occasional tumors of the liver, an organ that can incorporate dietary Neu5Gc. Neu5Gc-deficient mice immunized against Neu5Gc and fed bioavailable Neu5Gc developed a much higher incidence of hepatocellular carcinomas, with evidence of Neu5Gc accumulation. Taken together, our data provide an unusual mechanistic explanation for the epidemiological association between red meat consumption and carcinoma risk. This mechanism might also contribute to other chronic inflammatory processes epidemiologically associated with red meat consumption.


Asunto(s)
Inflamación/etiología , Neoplasias Hepáticas Experimentales/etiología , Carne/efectos adversos , Carne/análisis , Ácidos Neuramínicos/efectos adversos , Animales , Anticuerpos Bloqueadores/metabolismo , Progresión de la Enfermedad , Análisis de los Alimentos , Humanos , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Ácido N-Acetilneuramínico/análisis , Ácidos Neuramínicos/análisis , Ácidos Neuramínicos/inmunología , Factores de Riesgo
10.
Chembiochem ; 18(13): 1155-1171, 2017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28423240

RESUMEN

About 2-3 million years ago, Alu-mediated deletion of a critical exon in the CMAH gene became fixed in the hominin lineage ancestral to humans, possibly through a stepwise process of selection by pathogen targeting of the CMAH product (the sialic acid Neu5Gc), followed by reproductive isolation through female anti-Neu5Gc antibodies. Loss of CMAH has occurred independently in some other lineages, but is functionally intact in Old World primates, including our closest relatives, the chimpanzee. Although the biophysical and biochemical ramifications of losing tens of millions of Neu5Gc hydroxy groups at most cell surfaces remains poorly understood, we do know that there are multiscale effects functionally relevant to both sides of the host-pathogen interface. Hominin CMAH loss might also contribute to understanding human evolution, at the time when our ancestors were starting to use stone tools, increasing their consumption of meat, and possibly hunting. Comparisons with chimpanzees within ethical and practical limitations have revealed some consequences of human CMAH loss, but more has been learned by using a mouse model with a human-like Cmah inactivation. For example, such mice can develop antibodies against Neu5Gc that could affect inflammatory processes like cancer progression in the face of Neu5Gc metabolic incorporation from red meats, display a hyper-reactive immune system, a human-like tendency for delayed wound healing, late-onset hearing loss, insulin resistance, susceptibility to muscular dystrophy pathologies, and increased sensitivity to multiple human-adapted pathogens involving sialic acids. Further studies in such mice could provide a model for other human-specific processes and pathologies involving sialic acid biology that have yet to be explored.


Asunto(s)
Genoma , Pérdida Auditiva/metabolismo , Oxigenasas de Función Mixta/genética , Distrofias Musculares/metabolismo , Neoplasias/metabolismo , Ácidos Neuramínicos/metabolismo , Animales , Autoanticuerpos/biosíntesis , Evolución Biológica , Susceptibilidad a Enfermedades , Expresión Génica , Pérdida Auditiva/genética , Pérdida Auditiva/inmunología , Pérdida Auditiva/patología , Humanos , Resistencia a la Insulina , Ratones , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/inmunología , Distrofias Musculares/genética , Distrofias Musculares/inmunología , Distrofias Musculares/patología , Mutación , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Ácidos Neuramínicos/química , Ácidos Neuramínicos/inmunología , Pan troglodytes
11.
Mol Genet Metab ; 121(4): 329-335, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28673550

RESUMEN

Inborn defects of cholesterol biosynthesis are metabolic disorders presenting with multi-organ and tissue anomalies. An autosomal recessive defect involving the demethylating enzyme C4-methyl sterol (SC4MOL) has been reported in only 4 patients so far. In infancy, all patients were affected by microcephaly, bilateral congenital cataracts, growth delay, psoriasiform dermatitis, immune dysfunction, and intellectual disability. Herein, we describe a new case of SC4MOL deficiency in which a 19-year-old Italian male was affected by bilateral congenital cataracts, growth delay and learning disabilities, behavioral disorders and small stature, but not microcephaly. Our patient had abundant scalp dandruff, without other skin manifestations. Analysis of the blood sterol profile showed accumulation of C4-monomethyl and C4-dimethyl sterols suggesting a deficiency of the SC4MOL enzyme. Sequencing of the MSMO1 gene (also known as the "SC4MOL" gene) confirmed mutations in each allele (c.731A>G, p.Y244C, which is already known, and c.605G>A, p.G202E, which is a novel variant). His father carried c.731A>G mutation, whereas his mother carried c.605G>A. Thus, the combination of multiple skills and methodologies, in particular, blood sterol profiling and genetic analysis, led to the diagnosis of a new case of a very rare defect of cholesterol biosynthesis. Consequently, we suggest that these two analyses should be performed as soon as possible in all undiagnosed patients affected by bilateral cataracts and developmental delay.


Asunto(s)
Errores Innatos del Metabolismo Lipídico/diagnóstico , Errores Innatos del Metabolismo Lipídico/genética , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Esteroles/sangre , Alelos , Catarata/etiología , Colesterol/metabolismo , Discapacidades del Desarrollo/etiología , Familia , Humanos , Discapacidad Intelectual/etiología , Errores Innatos del Metabolismo Lipídico/sangre , Errores Innatos del Metabolismo Lipídico/complicaciones , Masculino , Microcefalia , Oxigenasas de Función Mixta/sangre , Mutación , Esteroles/química , Adulto Joven
12.
Biosci Biotechnol Biochem ; 81(8): 1536-1541, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28585465

RESUMEN

An elicitor chitosan (CHT) induces stomatal closure but the mechanism remains to be clarified. A phytohormone salicylic acid (SA) is crucial for elicitor-induced defense signaling in plants. Here we investigated whether endogenous SA is required for CHT signaling in guard cells. In the SA-deficient nahG mutant, treatment of CHT did not induce either apoplastic reactive oxygen species (ROS) production or stomatal closure but co-treatment of CHT and SA induced both apoplastic ROS production and stomatal closure, indicating the involvement of endogenous SA in CHT-induced apoplastic ROS production and CHT-induced stomatal closure. Furthermore, CHT induced transient cytosolic free calcium concentration increments in the nahG mutant in the presence of exogenous SA but not in the absence of exogenous SA. These results provide evidence that endogenous SA is a crucial element in CHT-induced stomatal closure.


Asunto(s)
Arabidopsis/efectos de los fármacos , Quitosano/farmacología , Reguladores del Crecimiento de las Plantas/metabolismo , Estomas de Plantas/efectos de los fármacos , Ácido Salicílico/metabolismo , Transducción de Señal , Ácido Abscísico/metabolismo , Arabidopsis/genética , Arabidopsis/metabolismo , Calcio/metabolismo , Expresión Génica , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Mutación , Células Vegetales/efectos de los fármacos , Células Vegetales/metabolismo , Reguladores del Crecimiento de las Plantas/farmacología , Estomas de Plantas/genética , Estomas de Plantas/metabolismo , Ácido Salicílico/farmacología
13.
Nat Chem Biol ; 10(7): 552-4, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24838010

RESUMEN

Despite the remarkable versatility displayed by flavin-dependent monooxygenases (FMOs) in natural product biosynthesis, one notably missing activity is the oxidative generation of carbonate functional groups. We describe a multifunctional Baeyer-Villiger monooxygenase, CcsB, which catalyzes the formation of an in-line carbonate in the macrocyclic portion of cytochalasin E. This study expands the repertoire of activities of FMOs and provides a possible synthetic strategy for transformation of ketones into carbonates.


Asunto(s)
Aspergillus/química , Carbonatos/química , Citocalasinas/química , Proteínas Fúngicas/química , Regulación Fúngica de la Expresión Génica , Cetonas/química , Oxigenasas de Función Mixta/química , Secuencia de Aminoácidos , Aspergillus/enzimología , Aspergillus/genética , Catálisis , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Técnicas de Inactivación de Genes , Prueba de Complementación Genética , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Oxidación-Reducción
14.
J Inherit Metab Dis ; 39(6): 821-829, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27488560

RESUMEN

Hawkinsinuria is a rare disorder of tyrosine metabolism that can manifest with metabolic acidosis and growth arrest around the time of weaning off breast milk, typically followed by spontaneous resolution of symptoms around 1 year of age. The urinary metabolites hawkinsin, quinolacetic acid, and pyroglutamic acid can aid in identifying this condition, although their relationship to the clinical manifestations is not known. Herein we describe clinical and laboratory findings in two fraternal twins with hawkinsinuria who presented with failure to thrive and metabolic acidosis. Close clinical follow-up and laboratory testing revealed previously unrecognized hypoglycemia, hypophosphatemia, combined hyperlipidemia, and anemia, along with the characteristic urinary metabolites, including massive pyroglutamic aciduria. Treatment with N-acetyl-L-cysteine (NAC) restored normal growth and normalized or improved most biochemical parameters. The dramatic response to NAC therapy supports the idea that glutathione depletion plays a key role in the pathogenesis of hawkinsinuria.


Asunto(s)
Acetilcisteína/uso terapéutico , Oxigenasas de Función Mixta/deficiencia , Tirosinemias/tratamiento farmacológico , Acidosis/patología , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Errores Innatos del Metabolismo de los Aminoácidos/patología , Femenino , Glutatión Sintasa/deficiencia , Humanos , Recién Nacido , Masculino , Fenotipo , Gemelos , Tirosinemias/patología
15.
Ann Nutr Metab ; 68 Suppl 3: 5-9, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27931018

RESUMEN

Carnitine is needed for transfer of long-chain fatty acids across the inner mitochondrial membrane for subsequent ß-oxidation. Carnitine can be synthesized by the body and is also obtained in the diet through consumption of meat and dairy products. Defects in carnitine transport such as those caused by defective activity of the OCTN2 transporter encoded by the SLC22A5 gene result in primary carnitine deficiency, and newborn screening programmes can identify patients at risk for this condition before irreversible damage. Initial biochemical diagnosis can be confirmed through molecular testing, although direct study of carnitine transport in fibroblasts is very useful to confirm or exclude primary carnitine deficiency in individuals with genetic variations of unknown clinical significance or who continue to have low levels of carnitine despite negative molecular analyses. Genetic defects in carnitine biosynthesis do not generally result in low plasma levels of carnitine. However, deletion of the trimethyllysine hydroxylase gene, a key gene in carnitine biosynthesis, has been associated with non-dysmorphic autism. Thus, new roles for carnitine are emerging that are unrelated to classic inborn errors of metabolism.


Asunto(s)
Cardiomiopatías/diagnóstico , Carnitina/deficiencia , Enfermedades Carenciales/diagnóstico , Pruebas Genéticas , Hiperamonemia/diagnóstico , Errores Innatos del Metabolismo/diagnóstico , Enfermedades Musculares/diagnóstico , Mutación , Tamizaje Neonatal , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Cardiomiopatías/dietoterapia , Cardiomiopatías/epidemiología , Cardiomiopatías/metabolismo , Carnitina/metabolismo , Carnitina/uso terapéutico , Enfermedades Carenciales/dietoterapia , Enfermedades Carenciales/metabolismo , Dinamarca/epidemiología , Suplementos Dietéticos , Humanos , Hiperamonemia/dietoterapia , Hiperamonemia/epidemiología , Hiperamonemia/metabolismo , Incidencia , Recién Nacido , Errores Innatos del Metabolismo/dietoterapia , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/metabolismo , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Enfermedades Musculares/dietoterapia , Enfermedades Musculares/epidemiología , Enfermedades Musculares/metabolismo , Pronóstico , Miembro 5 de la Familia 22 de Transportadores de Solutos/deficiencia , Miembro 5 de la Familia 22 de Transportadores de Solutos/metabolismo
16.
Infect Immun ; 83(3): 1039-47, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25547791

RESUMEN

Toxoplasma gondii infection has been described previously to cause infected mice to lose their fear of cat urine. This behavioral manipulation has been proposed to involve alterations of host dopamine pathways due to parasite-encoded aromatic amino acid hydroxylases. Here, we report successful knockout and complementation of the aromatic amino acid hydroxylase AAH2 gene, with no observable phenotype in parasite growth or differentiation in vitro and in vivo. Additionally, expression levels of the two aromatic amino acid hydroxylases were negligible both in tachyzoites and in bradyzoites. Finally, we were unable to confirm previously described effects of parasite infection on host dopamine either in vitro or in vivo, even when AAH2 was overexpressed using the BAG1 promoter. Together, these data indicate that AAH enzymes in the parasite do not cause global or regional alterations of dopamine in the host brain, although they may affect this pathway locally. Additionally, our findings suggest alternative roles for the AHH enzymes in T. gondii, since AAH1 is essential for growth in nondopaminergic cells.


Asunto(s)
Encéfalo/metabolismo , Estadios del Ciclo de Vida , Oxigenasas de Función Mixta/genética , Proteínas Protozoarias/genética , Toxoplasma/crecimiento & desarrollo , Toxoplasmosis Animal/metabolismo , Animales , Gatos , Dopamina/metabolismo , Femenino , Eliminación de Gen , Expresión Génica , Interacciones Huésped-Parásitos , Isoenzimas/deficiencia , Isoenzimas/genética , Ratones , Oxigenasas de Función Mixta/deficiencia , Plásmidos , Regiones Promotoras Genéticas , Proteínas Protozoarias/metabolismo , Toxoplasma/enzimología , Toxoplasma/genética , Toxoplasmosis Animal/parasitología
17.
Chembiochem ; 16(8): 1175-82, 2015 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-25907804

RESUMEN

The biologically active compound mensacarcin is produced by Streptomyces bottropensis. The cosmid cos2 contains a large part of the mensacarcin biosynthesis gene cluster. Heterologous expression of this cosmid in Streptomyces albus J1074 led to the production of the intermediate didesmethylmensacarcin (DDMM). In order to gain more insights into the biosynthesis, gene inactivation experiments were carried out by λ-Red/ET-mediated recombination, and the deletion mutants were introduced into the host S. albus. In total, 23 genes were inactivated. Analysis of the metabolic profiles of the mutant strains showed the complete collapse of DDMM biosynthesis, but upon overexpression of the SARP regulatory gene msnR1 in each mutant new intermediates were detected. The compounds were isolated, and their structures were elucidated. Based on the results the specific functions of several enzymes were determined, and a pathway for mensacarcin biosynthesis is proposed.


Asunto(s)
Antracenos/metabolismo , Luciferasas/genética , Oxigenasas de Función Mixta/genética , Familia de Multigenes/genética , Sistemas de Lectura Abierta/genética , Biocatálisis , Eliminación de Gen , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/aislamiento & purificación , Oxigenasas de Función Mixta/metabolismo , Streptomyces/enzimología , Streptomyces/genética , Streptomyces/metabolismo
18.
Top Curr Chem ; 366: 31-54, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-23471785

RESUMEN

The sialic acids N-acetylneuraminic acid (Neu5Ac) and N-glycolylneuraminic acid (Neu5Gc) differ by a single oxygen atom and are widely found at the terminal position of glycans on vertebrate cell surfaces. In animals capable of synthesizing Neu5Gc, most tissues and cell types express both sialic acids, in proportions that vary between species. However, it has long been noted that Neu5Gc is consistently expressed at trace to absent levels in the brains of all vertebrates studied to date. Although several reports have claimed to find low levels of Neu5Gc-containing glycans in neural tissue, no study definitively excludes the possibility of contamination with glycans from non-neural cell types. This distribution of a molecule - prominently but variably expressed in extraneural tissues but very low or absent in the brain - is, to our knowledge, unique. The evolutionarily conserved brain-specific suppression of Neu5Gc may indicate that its presence is toxic to this organ; however, no studies to date have directly addressed this very interesting question. Here we provide a historical background to this issue and discuss potential mechanisms causing the suppression of Neu5Gc expression in brain tissue, as well as mechanisms by which Neu5Gc may exert the presumed toxicity. Finally, we discuss future approaches towards understanding the mechanisms and implications of this unusual finding.


Asunto(s)
Encéfalo/metabolismo , Oxigenasas de Función Mixta/deficiencia , Ácido N-Acetilneuramínico/metabolismo , Ácidos Neuramínicos/metabolismo , Neuronas/metabolismo , Animales , Evolución Biológica , Química Encefálica , Membrana Celular/química , Membrana Celular/metabolismo , Gangliósidos/química , Gangliósidos/metabolismo , Expresión Génica , Humanos , Oxigenasas de Función Mixta/genética , Ácido N-Acetilneuramínico/química , Ácidos Neuramínicos/química , Neuronas/química , Especificidad de Órganos , Polisacáridos/química , Polisacáridos/metabolismo , Especificidad de la Especie
19.
Xenotransplantation ; 22(5): 379-90, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26381494

RESUMEN

BACKGROUND: Recent advancements in gene editing techniques have increased in number and utility. These techniques are an attractive alternative to conventional gene targeting methods via homologous recombination due to the ease of use and the high efficiency of gene editing. We have previously produced cytidine monophosphate-N-acetylneuraminic acid hydroxylase (CMAH) knockout (KO) pigs in a Minnesota miniature pig genetic background. These pigs were generated using zinc-finger nucleases (ZFNs) in combination with donor DNA containing a total homology length of 1600 bp (800-bp homology on each arm). Our next aim was to introduce the targeted disruption of alpha-1,3-galactosyltransferase (GGTA1) in the CMAH KO genetic background and evaluate the effect of donor DNA homology length on meganuclease-mediated gene targeting. METHODS: Zinc-finger nucleases from a previous CMAH KO experiment were used as a proof of concept to identify a correlation between the length of donor DNA homology and targeting efficiency. Based on those results, experiments were designed to use transcription activator-like effector nucleases (TALENs) to generate bi-allelically modified GGTA1 cells using donor DNAs carrying various lengths of homology. Donor DNA was designed to symmetrically flank the predicted cleavage sites in CMAH and GGTA1 for both ZFN and TALEN cleavage sites, respectively. For both genes, the length of total homology ranged from 60 to 1799 bp. Sialyltransferase gene expression profiles were evaluated in CMAH and GGTA1 double KO pig cells and were compared to wild-type and CMAH KO cells. RESULTS: Introduction of donor DNA with ZFNs demonstrated that small amounts of homology (60 bp) could facilitate homology-directed repair during ZFN-mediated targeting of CMAH; however, donor DNA with longer amounts of homology resulted in a higher frequency of homology-directed repair. For the GGTA1 KO experiments that used TALENs and donor DNA, donor DNA alone did not result in detectable bi-allelic conversion of GGTA1. As the length of donor DNA increased, the bi-allelic disruption of GGTA1 increased from 0.5% (TALENs alone, no donor DNA present) to a maximum of 3% (TALENs and donor DNA with total homology of 1799 bp). Inclusion of homologous donor DNA in TALEN-mediated gene targeting facilitated a higher incidence of bi-allelically modified cells. Using the generated cells, we were able to demonstrate the lack of GGTA1 expression and the decrease in gene expression sialyltransferase-related genes. CONCLUSIONS: The approach of using donor DNA in conjunction with a meganuclease can be used to increase the efficiency of gene targeting. The gene editing methods can be applied to other genes as well as other mammalian systems. Additionally, gene expression analysis further confirms that the CMAH/GGTA1 double KO pigs can be a valuable source for the study of pig-to-human xenotransplantation.


Asunto(s)
Animales Modificados Genéticamente , Marcación de Gen/métodos , Porcinos/genética , Alelos , Animales , ADN , Desoxirribonucleasas , Femenino , Galactosiltransferasas/deficiencia , Galactosiltransferasas/genética , Humanos , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Trasplante Heterólogo/métodos
20.
J Immunol ; 191(1): 228-37, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23709682

RESUMEN

Clinical evidence for a more active immune response in humans compared with our closest hominid relative, the chimpanzee, includes the progression of HIV infection to AIDS, hepatitis B- and C-related inflammation, autoimmunity, and unwanted harmful immune responses to viral gene transfer vectors. Humans have a unique mutation of the enzyme CMP-N-acetylneuraminic acid hydroxylase (CMAH), causing loss of expression of the sialic acid Neu5Gc. This mutation, occurring 2 million years ago, likely altered the expression and function of ITIM-bearing inhibitory receptors (Siglecs) that bind sialic acids. Previous work showed that human T cells proliferate faster than chimpanzee T cells upon equivalent stimulation. In this article, we report that Cmah(-/-) mouse T cells proliferate faster and have greater expression of activation markers than wild-type mouse T cells. Metabolically reintroducing Neu5Gc diminishes the proliferation and activation of both human and murine Cmah(-/-) T cells. Importantly, Cmah(-/-) mice mount greater T cell responses to an adenovirus encoding an adeno-associated virus capsid transgene. Upon lymphocytic choriomeningitis virus infection, Cmah(-/-) mice make more lymphocytic choriomeningitis virus-specific T cells than WT mice, and these T cells are more polyfunctional. Therefore, a uniquely human glycosylation mutation, modeled in mice, leads to a more proliferative and active T cell population. These findings in a human-like mouse model have implications for understanding the hyperimmune responses that characterize some human diseases.


Asunto(s)
Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Proliferación Celular , Células Cultivadas , Dependovirus/genética , Dependovirus/inmunología , Dependovirus/metabolismo , Glicosilación , Humanos , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxigenasas de Función Mixta/deficiencia , Oxigenasas de Función Mixta/genética , Subgrupos de Linfocitos T/enzimología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA